(19)
(11) EP 2 266 585 B1

(12) EUROPEAN PATENT SPECIFICATION

(45) Mention of the grant of the patent:
26.06.2013 Bulletin 2013/26

(21) Application number: 10185891.8

(22) Date of filing: 06.05.2004
(51) International Patent Classification (IPC): 
A61K 33/24(2006.01)
A61K 31/194(2006.01)
A61P 19/08(2006.01)
A61K 31/198(2006.01)

(54)

Water-soluble strontium salts for use in treatment of cartilage and/or bone conditions

Wasserlösliche Strontiumsalze zur Behandlung von Knorpel- und Knochenerkrankungen

Sels de strontium hydrosolubles pour le traitement des d'affections de cartilage et/ou d'os


(84) Designated Contracting States:
AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LI LU MC NL PL PT RO SE SI SK TR

(30) Priority: 07.05.2003 DK 200300691
20.06.2003 DK 200300932
09.12.2003 DK 200301820
09.12.2003 US 528442 P

(43) Date of publication of application:
29.12.2010 Bulletin 2010/52

(62) Application number of the earlier application in accordance with Art. 76 EPC:
06021612.4 / 1745791
04731317.6 / 1534305

(73) Proprietor: Osteologix A/S
2100 Copenhagen (DK)

(72) Inventors:
  • Christgau, Stephan
    DK-2820, Gentofte (DK)
  • Hansen, Christian
    DK-2950, Vedbæk (DK)
  • Andersen, Jens
    DK-2950, Vedbæk (DK)
  • Nilsson, Henrik
    8835 Feusisberg (CH)

(74) Representative: Hjelmencrantz, Anders et al
Chas. Hude A/S H.C. Andersens Boulevard 33
1780 Copenhagen V
1780 Copenhagen V (DK)


(56) References cited: : 
EP-A1- 0 813 869
US-A- 5 856 356
GB-A- 2 345 060
US-A- 6 139 850
   
  • SORBERA L A ET AL: "STRONTIUM RANELATE TREATMENT AND PREVENTION OF OSTEOPOROSIS BONE RESORPTION INHIBITOR BONE FORMATION STIMULANT", DRUGS OF THE FUTURE, BARCELONA, ES, vol. 28, no. 4, 1 April 2003 (2003-04-01), pages 328-335, XP008036784, ISSN: 0377-8282
  • GASTINEAU G F ET AL: "METABOLIC STUDIES OF A PATIENT WITH OSTEOPOROSIS AND DIABETES MELLITUS: EFFECTS OF TESTOSTERONE ENANTHATE AND STRONTIUM LACTATE", MAYO CLINIC PROCEEDINGS, DOWDEN HEALTH MEDIA, INC, US, vol. 35, 2 March 1960 (1960-03-02), page 105, XP008036793, ISSN: 0025-6196
  • REGINSTER J-Y ET AL: "STRONTIUM RANELATE: A NEW PARADIGM IN THE TREATMENT OF OSTEOPOROSIS", DRUGS OF TODAY / MEDICAMENTOS DE ACTUALIDAD, J.R. PROUS SS.A. INTERNATIONAL PUBLISHERS, ES LNKD- DOI:10.1358/DOT.2003.39.2.799416, vol. 39, no. 2, 1 February 2003 (2003-02-01), pages 89-101, XP008036782, ISSN: 0025-7656
  • GHANDOUR M A: "pH-metric studies of mixed-ligand complexes of Cadmium-, Magnesium-, Strontium- and Calcium-(II) with pyruvate and oxalate or citrate", JOURNAL OF THE INDIAN CHEMICAL SOCIETY, THE INDIAN CHEMICAL SOCIETY, CALCUTTA; IN, vol. 65, 1 October 1988 (1988-10-01), pages 716-718, XP008133220, ISSN: 0019-4522
 
Remarks:
The file contains technical information submitted after the application was filed and not included in this specification
 
Note: Within nine months from the publication of the mention of the grant of the European patent, any person may give notice to the European Patent Office of opposition to the European patent granted. Notice of opposition shall be filed in a written reasoned statement. It shall not be deemed to have been filed until the opposition fee has been paid. (Art. 99(1) European Patent Convention).


Description

Field of the Invention



[0001] The present invention relates to compounds and pharmaceutical compositions for use in the treatment and/or prophylaxis of specific cartilage and/or bone conditions

[0002] The present invention also relates to an improved method for preparing the strontium salt of glutamic acid.

Background of the invention



[0003] Osteoporosis is the most common form of metabolic bone disease in humans. It is a condition, which affects a very large number of people all over the world, and as the number of elderly people is set to rise dramatically in the coming decades in most countries, the prevalence and impact of osteoporosis will also increase. The disease is characterized pathologically by an absolute decrease in the amount of bone mass and the structural quality of bone, and clinically by increased susceptibility to fractures. In fact, osteoporosis is the most significant underlying cause of skeletal fractures in late middle aged and elderly women.

[0004] In general, there are two types of osteoporosis: primary and secondary. Secondary osteoporosis is the result of an identifiable disease process or agent. However, approximately 90% of all osteoporosis cases are idiopathic primary osteoporosis. Such primary osteoporosis includes postmenopausal osteoporosis, age-associated osteoporosis (affecting a majority of individuals over the age of 70 to 80), and idiopathic osteoporosis affecting middle-aged and younger men and women.

[0005] The mechanism of bone loss in osteoporosis is believed to involve an imbalance in the process of bone remodeling. Bone remodeling occurs throughout life, renewing the skeleton and maintaining the strength of bone. This remodeling is mediated by specialized cells of the bone tissue, called "osteoclasts" and "osteoblasts". Osteoclasts (bone dissolving or resorbing cells) are responsible for the resorption of a portion of bone within the bone matrix, during the resorption process. After resorption, the osteoclasts are followed by the appearance of osteoblasts (bone forming cells), which then refill the resorbed portion with new bone.

[0006] The formation of the two cell types as well as their activity in bone is usually tightly coupled and well regulated in order to maintain the skeletal balance and structural integrity of the bones. However, in people with osteoporosis an imbalance in this remodeling process develops, resulting in loss of bone at a rate faster than the accretion of bone.

[0007] The single most important risk factor for osteoporosis is oestrogen deficiency occurring naturally at the menopause. The decline in endogenous oestrogen production leads to an elevated metabolic activity in the bone tissue where the increase in osteoclast mediated bone resorption surpasses the more modest increase in bone formation resulting in a net loss of bone. The actual number of people affected will grow at a rate greater than simple population growth rates, because the aging of the population is disproportionately increasing the older segment of the population, while the age for the onset of menopause has remained constant. In the last decades there has also been a substantial advance in the ability to predict and monitor osteoporosis, as methods for measurement of bone mineral density (BMD) has improved and new specific biochemical markers of bone resorption and formation has been developed and made available for routine clinical use. New pharmaceutical agents for treatment and/or prevention of osteoporosis have also been developed. The majority of these treatments are either based on substituting the lost endogenous estrogen either in the form of hormone replacement therapy (HRT) or selective estrogen receptor modulators (SERM), or they belong to the class of compounds called bisphosphonates. SERM's and especially HRT is associated with significant side effects, such as increased risk of cancer and cardiovascular disease, whereas bisphosphonates in addition to a potent antiresorptive effect also decreases bone formation to a similar extent, implying that they loose their therapeutic effect after few years of treatment. Thus, there is a need for agents, which are effective in the treatment and/or prophylaxis of osteoporosis.

[0008] SORBERA LA ET AL: "STRONTIUM RANELATE TREATMENT AND PREVENTION OF OSTEOPOROSIS BONE RESORPTION INHIBITOR BONE FORMATION STIMULANT", DRUGS OF THE FUTURE, PROUSSCIENCE, E5, vol. 28, no. 4, 1 April 2003 (2003-04-01), pages 328-335, discloses the use of a soluble strontium salt of an organic acid (strontium ranelate, a synthetic non-naturally occurring thiophene-substituted amino acid) for the treatment of osteoporosis. In the studies discussed in said document. PREVOS and STRATOS, Replace with "Stromtium ranelate was administered" once a day see table I, 1000 mg perorally, once daily. Said document furthermore describes that strontium ranelate has good bioavailability. Said document is therefore the closest prior art.

Description of the invention



[0009] The scope of the invention is limited by the spended claims.

[0010] Previous studies have shown that various strontium compounds modulate bone loss in osteoporosis when present at levels higher than those required for normal cell physiology. The effect is believed to be due to a stimulatory effect of strontium on pre-osteoblastic cell differentiation and migration, and a direct or matrix-mediated inhibition of osteoclast activity by strontium (Reginster, JY, Curr pharm Des 2002:8 (21):1907-16). In other words, strontium both works as an anti-resorptive and an anabolic agent. Various salts of strontium are known from the prior art, such as, e.g., strontium ranelate (distrontium salt of 2-[N,N-di(carboxymethyl)amino]-3-cyano-4-carboxymethylthiophene-5-carboxylic acid) described in EP-B 0 415 850. The ranelate part of the strontium compound, derived from ranelic acid, is unlikely to have any therapeutic effect towards cartilage or bone conditions per se. Other known strontium salts are e.g., strontium tartrate, strontium phosphate, strontium carbonate, strontium nitrate, strontium sulfate and strontium chloride.

[0011] The naturally occurring salts of strontium, such as the carbonate and sulphate salts, have very low water solubility (0.15 g/l or below at room temperature). In contrast, the other strontium salts, such as strontium chloride, strontium hydroxide, strontium nitrate, strontium oxide and strontium acetate have very high solubilities in the range from 225 - 800 g/l in water. In this respect the strontium salts are very similar to the corresponding magnesium and calcium salts.

[0012] Organic strontium salts have been described, but literature reports of this type of compounds are limited to rather few substance. Again in these cases the physiochemical properties have been reported to be very similar to the corresponding magnesium, calcium and barium salts. Carboxylic acids can form stable crystalline salts with divalent earth metals such as strontium, and especially di-carboxylic acids are interesting, as they can have a partial chelating effect. Such complexation may be important in biological systems, where the alkaline earth metals, especially calcium and magnesium, plays important physiological roles (). Hence the divalent metal ions may exist in a complex form in the aqueous environment in biological systems, rather than in a free and un-bound ionic form. Complex formation constants with the alkaline earth metals in aqueous solution are higher for amino acids than for hydroxy-carboxylic acids and the related non-carboxylic acids, which suggest that the amino group may play a role in the complex formation. Generally the differences in association constants for the various ligands becomes smaller as the radius of the metal increases, and thus stability of strontium complexes with di-carboxylic acid is lower than the stability of the comparable complexes with calcium and magnesium.

[0013] For a pharmaceutical application of the strontium salts this is very important as it means that strontium salts of dicarboxylic amino acids may be particularly useful. We have found that such salts, such as strontium glutamate and strontium aspartate are more soluble than other dicarboxylic strontium salts of similar molecular size. In pure aqueous solutions of such salts strontium exists in partly complexed form. However, when administered to an animal such as a mammal, i.e. a rat, dog, monkey or human, ionic strontium as well as strontium complexed to the carboxylic acid anion will be taken up from the intestinal lumen by both passive and active transport mechanisms. In this case strontium will be displaced from the complexes by available calcium and magnesium which forms much more stable complexes with the ionized amino acids. It appears that with the heavy group II metals, such as strontium, the amino group in both aspartate and glutamate is much less significant for metal complexation, probably due to the unfavourable chelation of large metals in five- or six-membered rings. Accordingly, dianionic amino-acid salts of strontium, such as strontium aspartate and strontium glutamate may be especially suited for prophylactic and/or therapeutic interventions in bone disease as the amino-acids may act to preferentially bind/complex with available free calcium, thus promoting both the intestinal uptake of the calcium ion, and physiological action of the ion, in particular its role in regulation of bone turnover.

[0014] The known strontium salt that are water-soluble have a water-solubility of at least about 225-800 g/l and the other known strontium salts have solubilities that are very low (below 0.1g/l at room temperature).

[0015] The present invention relates to strontium salts and pharmaceutical compositions containing them for use in the treatment and/or prophylaxis of cartilage and/or bone conditions

[0016] Also disclosed is that for the-treatment and/or prophylaxis of a cartilage and/or bone disease and/or conditions resulting in a dysregulation of cartilage and/or bone metabolism in a mammal, such as, e.g., a human female or male adult, adolescent or a child, such as, e.g., osteoporosis, osteoarthritis, osteopetrosis, osteopenia and Paget's disease, hypercalcemia of malignancy, periodontal disease, hyperparathyroidism, periarticular erosions in rheumatoid arthritis, osteodystrophy, myositis ossificans, Bechterew's disease, malignant hypercalcemia, osteolytic lesions produced by bone metastasis, bone pain due to bone metastasis, bone loss due to sex steroid hormone deficiency, bone abnormalities due to steroid hormone treatment, bone abnormalities caused by cancer therapeutics, osteomalacia, Bechet's disease, hyperostosis, metastatic bone disease, immobilization-induced osteopenia or osteoporosis, or glucocorticoid-induced osteopenia or osteoporosis, osteoporosis pseudoglioma syndrome, idiopathic juvenile osteoporosis, for the improvement of fracture healing after traumatic or atraumatic fracture, and for the maintenance or increase of energy level, for building up or strengthening muscle tissues and for weight gain, the present inventors have found that the use of a strontium salt has prophylactic and/or therapeutic value in that one or more of the following beneficial effects can be obtained:
  1. i) an improved bioavailability of strontium,
  2. ii) an improved absorption of strontium,
  3. iii) a reduction of side effects,
  4. iv) a flexible dose adjustment of strontium in order to tailor prevention and/or treatment, of a specific disease stage,
  5. v) a possible reduction in daily dosage
  6. vi) a possible reduction of the number of different pharmaceutical compositions that a patient must use to achieve a therapeutic effect.


[0017] Suitable strontium salts for use according to the invention or for use in admixture with a strontium salt according to the invention are found in the following list. However, only those salts that have a water-solubility of at least about 1 g/l and at the most about 100 g/l are object of the present invention. Such strontium salts are the amino acid salts strontium glutamate and strontium aspartate; strontium pyruvate, strontium alpha-ketoglutarate, strontium maleate and strontium succinate.

[0018] The organic acid may be selected from the group consisting of specific amino and other organic acids, i.e. maleic acid, pyruvic acid, L- and D-aspartic acid, L- and D-glutamic acid, alpha-ketoglutaric acid, and succinic acid.

[0019] The strontium salt for use according to the invention is water soluble, having a water solubility of at least 1 g/l, such as, e.g., at least 5 g/l, at least 10 g/l, at least 20 g/l, at least 30 g/l, at least 40 g/I, at least 50 g/l, at least 60 g/l, at least 70 g/I, at least 80 g/I, at least 90 g/l or about 100 g/I measured at room temperature, i.e a temperature of 20-25°C.

[0020] Specific examples of strontium salts for use according to the invention are strontium succinate, strontium aspartate in either L and/or D-form, strontium glutamate in either L- and/or D-form, strontium pyruvate, strontium maleate, and mixtures thereof.

[0021] Strontium L-glutamate (hexahydrate) has previously been prepared by reacting strontium hydroxide with L-glutamic acid under reflux for 3 hours with a subsequent cooling and slow crystallization over a period of 2 weeks. The crystals were subjected to X-ray crystallography in order to elucidate the crystal structure (please see: H. Schmidbaur, I. Bach, L. Wilkinson & G. Müller (1989), Chem Ber. 122; 1433-1438). The investigations were related to a crystalline form of the strontium salts with the properties described in figures 1 and 2 and tables 2 and 3.
  Distances  
Sr - O1 2.724(2) Sr - O2 2.665(2)
Sr - O1' 2.642(2) Sr - O2' 2.651(2)
Sr - O3' 2.677(2) Sr - O4' 2.658(2)
Sr - O5 2.658(2) Sr - O6 2.708(2)
Sr - O7 2.640(2) O1 - C1 1.268(3)
O2 - C1 1.258(3) C1 - C2 1.521(3)
C2 - N 1.469(3) C2 - C3 1.526(3)
C3 - C4 1.524(4) C4 - C5 1.513(4)
C5 - O3 1.264(3) C5 - O4 1.264(3)
  Angles  
O1 -Sr- O1' 115.4(1) O1 -Sr- O2 48.4(1)
O1 -Sr- O2' 67.8(1) O1 -Sr- O3' 75.4(1)
O1 -Sr- O4' 120.8(1) O1 -Sr- O5 74.8(1)
O1 -Sr- O6 105.7(1) O1 -Sr- O7 146.5(1)
O2 -Sr- O1' 68.8(1) O2 -Sr- O2' 115.3(1)
Table 2: Distances [A] and angles [°] for strontium L-glutamate hexahydrate as described by Schmidbaur et al 1989. For atomic numbering please see figure 1. For preparing symmetry operations 01' was derived from 01 by the operation: 0.5 + X, 0.5 X, -Z; 02' was derived from 02 by the operations X -0.5, 0.5 -Y, -Z; and 03' and 04' was derived from 03 and 04 respectively by the operation X, Y-1, Z. The parenthesis indicates the estimated units of the last significant figure.
O2 -Sr- O3' 79.3(1) O2 -Sr- O4' 122.1(1)
O2 -Sr- O5 98.4(1) O2 -Sr- O6 76.8(1)
O2 -Sr- O7 154.8(1) O2' -Sr- O1' 153.8(1)
O2' -Sr- O3' 75.5(1) O2' -Sr- O4' 78.9(1)
O2' -Sr- O5 70.9(1) O2' -Sr- O6 138.1(1)
O2' -Sr- O7 86.7(1) O3' -Sr- O1' 80.4(1)
O3' -Sr- O4' 48.8(1) O3' -Sr- O5 141.3(1)
O3' -Sr- O6 145.3(1) O3' -Sr- O7 120.2(1)
O4' -Sr- O1' 77.7(1) O4' -Sr- O5 137.2(1)
O4' -Sr- O6 130.7(1) O4' -Sr- O7 72.0(1)
O1' -Sr- O5 135.2(1) O1' -Sr- O6 67.7(1)
O1' -Sr- O7 97.1(1) O7 -Sr- O5 76.5(1)
O7 -Sr- O6 78.5(1) O6 -Sr- O5 67.6(1)
O1 - C1 - O2 122.1(2) O1 - C1 - C2 119.7(3)
O2 - C1 - C2 118.2(3) C1 - C2 - N 114.5(2)
N - C2 - C3 111.1(2) C1 - C2 - C3 109.9(2)
C2 - C3 - C4 114.5(2) C3 - C4 - C5 114.1(3)
C4 - C5 - O3 119.7(2). C4 - C5 - O4 118.7(2)
O3 - C5 - O4 121.5(2)    
Table 3: Fractional atomic coordinates and equivalent isotropic thermal parameters for strontium L-glutamate hexahydrate as described by Schmidbaur et al 1989. Ueq= (U1*U2*U3), where U1, U2 and U3 are the intrinsic values of the Uji matrix. For atom nomenclature please refer to figure 1.
Atom X/A Y/B Z/C U(eq.)
Sr 0.9078 0.1999 0.0562 0.016
O1 0.7595 0.3566 -0.0465 0.023
O2 1.0591 0.3655 -0.0393 0.022
C1 0.9123 0.4069 -0.0655 0.017
C2 0.9206 0.5254 -0.1202 0.019
N 0.7562 0.5338 -0.1612 0.031
C3 0.9679 0.6810 -0.0913 0.024
C4 0.8471 0.7306 -0.0342 0.033
C5 0.8953 0.8849 -0.0059 0.021
O3 0.9030 0.9998 -0.0434 0.024
O4 0.9172 0.8970 0.0557 0.026
O5 0.7071 0.4172 0.1114 0.029
O6 1.1116 0.4051 0.1232 0.030
O7 0.8664 0.1049 0.1788 0.034
O8 0.3894 -0.1997 0.2655 0.042
O9 0.9133 -0.3339 0.1451 0.033
010 0.7665 -0.1770 0.2495 0.047


[0022] As apparent from the data disclosed in figures 1 and 2 and table 2 and 3, the strontium glutamate salt in hexahydrate form described by Scmidbaur et al is an orthorhombic crystal form belonging to space group P212121. The cell size is defined by the following dimensions (given in A): a 3.355, b 8.772, c 20.283, with a unit cell volume of 1308.6 Å3. The solubility of the isolated strontium glutamate (hexahydrate) with the properties as described (Schmidbaur, I Bach, L Wilkinson & G Müller (1989), Chem Ber. 122; 1433-1438) was reported as 0.023 g/l at 20°C.

[0023] Strontium L-aspartate has also previously been prepared by reacting L-aspartic acid with strontium hydroxide. The reaction was performed over 3 hours under reflux, and the resulting reaction mixture was allowed to cool over three days to initiate crystal formation. The resulting strontium L-aspartate crystals were subjected to X-ray crystallography in order to elucidate the crystal structure (please see: H. Schmidbaur, P. Mikulcik & G. Müller (1990), Chem Ber. 123; 1599-1602). The investigations revealed that the isolates strontium L-aspartate salt was formed in the trihydrate form with the properties described in figure 3 and tables 4 and 5.
  Distances  
Sr - O1 2.713(4) Sr - O2 2.707(5)
Sr - O3" 2.666(6) Sr - O3" 2.635(8)
Sr - O4" 2.799(7) Sr - O4"' 2.580(7)
Sr - O5 2.568(8) Sr - O6 2.683(7)
Sr - O7 2.627(3) O1 - C1 1.258(7)
O2 - C1 1.254(7) C1 - C2 1.540(8)
C2 - C3 1.510(9). C3 - C4 1.522(7)
O3 - C4 1.29(1) O4 - C4 1.23(1)
  Angles  
O1 -Sr- O2 48.0(1) O1 -Sr- O3' 84.2(2)
O2 -Sr- O3' 88.5(2) O3" -Sr- O3' 112.4(2)
O4" -Sr- O3' 65.4(2) O5 -Sr- O3' 70.3(2)
O6 -Sr- O3' 140.5(2) O3' -Sr- O4"' 152.7(2)
O3' -Sr- O7 73.3(2) O1 -Sr- O3" 146.9(2)
O2 -Sr- O3" 152.6(2) O3" -Sr- O4" 48.1(1)
O3" -Sr- O5 97.1 (2) O3" -Sr- O6 78.0(2)
O3" -Sr- O7 80.8(1) O1 -Sr- O4" 144.2(2)
O2 -Sr- O4" 141.9(2) O4" -Sr- O5 72.6(2)
O4" -Sr- O6 107.6(2) O4" -Sr- O7 76.6(2)
O1 -Sr- O4"' 83.3(2) O2 -Sr- O4"' 100.8(2)
O3' -Sr- O4"' 152.7(2) O3" -Sr- O4"' 69.0(2)
O4" -Sr- O4"' 115.2(2) O5 -Sr- O4"' 137.0(2)
O6 -Sr- O4"' 66.6(2) O7 -Sr- O4"' 80.3(2)
O1 -Sr- O6 107.9(2) O2 -Sr- O6 74.6(2)
O5 -Sr- O6 70.7(1) O1 -Sr- O7 76.9(1)
O1 -Sr- O5 115.7(2) O2 -Sr- O5 72.7(2)
O2 -Sr- O7 123.7(1) O5 -Sr- O7 139.5(2)
O6 -Sr- O7 145.5(2) O1 - C1 - O2 122.5(5)
O1 - C1 - C2 118.2(5) O2 - C1 - C2 119.1(5)
N - C2 - C1 116.3(5) N - C2 - C3 111.4(6)
C1 - C2 - C3 109.9(5) C2 - C3 - C4 115.2(6)
O3 - C4 - O4 123.8(5) O3 - C4 - C3 117(1)
O4 - C4 - C3 119(1)    


[0024] Table 3: Distances [Å] and angles [°] for strontium L-aspartate trihydrate as described by Schmidbaur et al 1990. For atomic numbering please see figure 3. The parenthesis indicates the estimated units of the last significant figure.
Table 4: Fractional atomic coordinates and equivalent isotropic thermal parameters for strontium glutamate hexahydrate as described by Schmidbaur et al 1990. Ueq = (U1*U2*U3), where U1, U2 and U3 are the intrinsic values of the Uji matrix. For atom nomenclature please refer to figure 3.
Atom X/A Y/B Z/C U(eq.)
Sr 0.2512 0.12345 0.01532 0.022
O1 0.247 -0.1101 -0.1041 0.046
O2 0.1997 -0.1361 0.0783 0.039
O3 0.3983 -0.6359 0.0410 0.049
O4 0.0957 -0.6194 0.0327 0.040
O5 0.0536 0.1264 0.1947 0.059
O6 0.4661 0.0865 0.1965 0.033
O7 0.238 0.2068 -0.1951 0.039
N 0.230 -0.3876 -0.1511 0.037
C1 0.2138 -0.1831 -0.0196 0.038
C2 0.1785 -0.3343 -0.0395 0.036
C3 0.263 -0.4160 0.0549 0.046
C4 1.1116 -0.5682 0.0416 0.034


[0025] As apparent from the data disclosed in figures 3 and table 3 and 4, the strontium glutamate salt in hexahydrate form described by Scmidbaur et al is an orthorhombic crystal form belonging to space group P212121. The cell size is defined by the following dimensions (given in A): a 7.304, b 9.914, c 11.837, with a unit cell volume of 857.1 Å3. The solubility of the isolated strontium aspartate trihydrate salt was not reported (H. Schmidbaur, P. Mikulcik & G. Müller (1990), Chem Ber. 123; 1599-1602).

[0026] Other examples of relevant acids for making strontium salts for use in a pharmaceutical composition may be found in WO 00/01692, which is hereby incorporated by reference.

Synthesis of strontium salts This section on systhesis does not form part of the invention.



[0027] Organic strontium salts of carboxylic acid anions can be synthesized by a number of different pathways. A conventional method for preparation of such organic strontium salts is to utilise the reaction between and organic acid and strontium hydroxide in an aqueous solution. This neutralisation reaction of, e.g. fumaric acid and strontium hydroxide salt follows the following scheme:

        Sr2+(aq)+ 2OH-(aq)+ HOOCCHCHCOOH(aq)→ Sr(OOCCHCHCOO)(aq)+ 2H2O(l)



[0028] The suspension of dissolved strontium fumarate can then be induced to precipitate by sublimation of water and subsequent up-concentration of the salt. Crystals will slowly form and precipitate from the solution.

[0029] An alternative approach is to utilise the sodium or potassium salt of the appropriate carboxylic acid anion and strontium chloride. As all organic strontium salts will be less soluble than the highly soluble chloride salt, the organic strontium salt will precipitate under these conditions leaving NaCl and excess SrCl2 in the solution. The equation below exemplifies this reaction scheme using as an example the reaction between SrCl2 and sodium-fumarate.

        Sr2+(aq) + 2Cl- (aq)+ 2Na+ (aq) + C4H2O42- (aq) → Sr(OOCCHCHCOO)(aq)+ Cl-(aq)+Na+(aq)



[0030] The present inventors have found that different strontium salts requires different synthesis pathways, and for some strontium salts we have identified optimized synthesis and manufacturing procedures. It has been found that synthesis of strontium salts of the di-carboxylic aminoacids aspartate and glutamate (in either D- or L- form) is very difficult when following these conventional reaction pathways, and generally results in low yields and purity of the obtained crystalline salt. In order to facilitate large-scale manufacture of pure strontium salts of dicarboxylic amino acids to carry out the pharmaceutical use according to the present invention, the present inventors have studied various synthesis pathways of these particular strontium salts. Thus, it has surprisingly been found that synthesis of well defined and pure strontium glutamate in hexahydrate form is most convenient carried out with the free acid form of glutamate and strontium hydroxide and requires elevated temperatures, such as temperatures above 80°C, or more preferred 100°C or even 120°C or most preferred more than 130°C Furthermore, we have found that addition of small volumes of alcohol can accelerate the crystal-formation of dissolved aqueous organic strontium salts. The inventors have also found that synthesis of strontium L-glutamate from L-glutamic acid and SrCl2 results in a hexahydrarte crystalline form distinct from the previously described strontium L-glutamate hexahydrate

[0031] Examples of these synthesis procedures for organic strontium salts of relevance for the treatment and/or prophylaxis of specific bone disease are provided in the examples herein.

[0032] A method for the preparation of strontium salts including strontium glutamate is described herein.

[0033] As mentioned above strontium is believed to have an effect on cartilage and/or bone conditions and/or other conditions, thus the salt may be used for the preparation of a pharmaceutical composition for the treatment and/or prophylaxis of a cartilage and/or a bone condition including the ones claimed . The pharmaceutical composition may further comprise one or more physiologically acceptable excipients.

[0034] For the treatment and/or prophylaxis of a cartilage and/or bone disease and/or conditions as claimed resulting in a dysregulation of cartilage and/or bone metabolism in a mammal, the possibility of administering various amounts of strontium in the form of one of the claimed salts and, if relevant alpha-ketoglutarate or an amino acid like e.g. glutamic acid and/or aspartic acid, respectively, may be desired. The amount of strontium (and, if relevant e.g.alpha-ketoglutarate or an amino acid) in a pharmaceutical composition according to the invention may be adjusted by adding an additional amount of strontium in the form of a strontium-containing compound (and/or, if relevant, an additional amount of alpha-ketoglutarate or an amino acid) to the composition. The strontium-containing compounds is selected from the salts claimed.

[0035] In certain cases it may be beneficial to further add one or more active substances to a pharmaceutical composition according to the invention. The one or more active substances may have a therapeutic and/or prophylactic effect on a cartilage and/or a bone disease and/or other conditions such as those mentioned above. The term "active substance having a therapeutic and/or prophylactic effect on diseases and conditions affecting metabolism and structural integrity of cartilage and/or bone" includes active substances that can attain a particular medical result, such as, e.g., reduce the incidence of bone fracture, increase bone density and/or improve healing of bone or decrease or halt the degradation of articular cartilage or promote formation of new cartilage or prevent or decrease progression of radiological evident joint damage. Examples of such substances are bone anti-resorptive and/or anabolic agents. However, one or more active substances having other effects than those mentioned above may also be included in a pharmaceutical composition of the invention. Such active substances could be e.g. disease-modifying anti-rheumatic drugs, or other anti-rheumatic drugs.

[0036] Specific examples of active substances, which may be used in a pharmaceutical composition according to invention are calcium-alpha-ketoglutarate, calcium and/or salts thereof, vitamin D such as, e.g., vitamin D3 and/or functional equivalents of vitamin D3, glucagon-like peptide-2, glucagon-like peptide-2 releasing compositions, bisphosphonates including ibandronate, zoledronate, alendronate, risedronate, ethidronate chlodronate, tiludronate and pamidronate; selective estrogen receptor modulators (SERMs) including raloxifene, arzoxifene, droloxifene, tamoxifen, 4-hydroxy-tamoxifen, 4'-iodotamoxifen, toremifene, (deaminohydroxy)-toremifene, chlomiphene, levormeloxifene, ormeloxifene, chroman derivatives, coumarin derivatives, idoxifene, nafoxidine, TAT-59, LY-353381, CP-336156, MDL-103323, EM-800, ICI-182,ICI 183,780, ICI 164,384, ICI 183,780, ICI 164,384, diethylstilbesterol, genistein, nafoxidine, nitromifene citrate, moxesterol, diphenol hydrochrysene, erythro-MEA, allenolic acid, equilin-3-sulphate, cyclophenyl, chlorotrianisene, ethamoxytriphetol, lasofoxifene, bazedoxifene, genistein, tibolone, ospemifene, tesmilffene, droloxifene, panomifene, zindoxifene, meproxifene and faslodex; calcitonin, parathyroid hormone, parathyroid hormone related peptide, glucosamine sulphate, glutamic acid and/or salts thereof, aspartic acid and/or salts thereof, proline, glutamine and hydroxyproline.

[0037] As mentioned above, the compounds and compositions of the present invention may be used for the treatment and prophylaxis of various conditions. Thus, also disclosed is a method for the treatment and/or prophylaxis of a cartilage and/or bone disease and/or conditions resulting in a dysregulation of cartilage and/or bone metabolism in a mammal, such as the ones mentioned above, for the maintenance or increase of energy level, for building up or strengthening muscle tissues and for weight gain, the method comprising administering to a subject in need thereof a therapeutic and/or prophylactic effective amount of a strontium salt having a water-solubility in the range claimed herein.

[0038] The subject is a human female or male adult, adolescent, or a child.

[0039] Also disclosed is a method for the treatment and/or prophylaxis of a cartilage and/or bone disease and/or conditions resulting in a dysregulation of cartilage and/or bone metabolism in a mammal, such as the ones mentioned above, for the maintenance or increase of energy level, for building up or strengthening muscle tissues and for weight gain, the method comprising administering to a subject in need thereof an amount of the strontium salt according to the invention.

[0040] The daily dose of strontium for use in the invention may be at least about 0.01 g, such as, e.g., at least about 0.025 g, at least about 0.050 g, at least about 0.075 g, at least about 0.1 g, at least about 0.2 g, at least about 0.3 g, at least about 0.4 g or at least about 0.5 g or from about 0.01 g to about 2 g such as, e.g., from about 0.1 g to about 2 g, from about 0.3 g to about 2 g or from about 0.3 g to about 1 g.

[0041] The invention also relates to compounds for use as claimed in a method wherein the strontium salt is administered in the form of a pharmaceutical composition as described above.

[0042] The invention further relates to compounds for use as claimed in a method wherein the administration may take place one or more times daily, such as from 1 to 5 times daily.

[0043] The invention also relates to compounds for use as claimed in a method wherein the administration may take place one or more times weekly, such as from 1 to 3 times weekly.

[0044] As described above, one or more active substances may be added to a pharmaceutical composition according to the invention, or administered as part of the same treatment as the administration of strontium salt. One example of such an active substance is Vitamin D. Vitamin D plays a major role in calcium absorption, since activated vitamin D3 (1,25-dihydroxycholecalciferol) and to a smaller extent other active forms of vitamin D, acts to increase the calcium absorption from the small intestine. Vitamin D3 acts to increase the entry of calcium through the plasma membrane into the enterocytes, and is capable of reducing the excretion of calcium to urine by increasing the reabsorbtion of calcium in kidneys. It is likely that vitamin D has the same effect on strontium absorption as it has on calcium absorption.

[0045] Vitamin D is activated in e.g. the liver and kidneys. High levels of calcium are having a reducing effect on activation of vitamin D, and high levels of strontium will probably have the same effect as calcium on the activation of vitamin D.

[0046] Thus, the administration of an amount of vitamin D together with a strontium-containing compound according to the invention will most likely have a beneficial effect on the uptake of strontium.

[0047] Accordingly, the invention relates to a use according to the invention further comprising administering a daily dose of vitamin D.

[0048] In a specific embodiment, vitamin D may be vitamin D3, and the daily dose of vitamin D3 may be at least about 1 µg, such as, e.g. at least about 1.25 µg at least about 1.50 µg, at least about 2 µg, at least about 3 µg, at least about 4 µg, at least about 5 µg, at least about 10 µg, at least about 15 µg, at least about 20 µg, at least about 25 µg, at least about 30 µg, at least about 40 µg or at least about 50 µg or from about 1 µg to about 50 µg such as, e.g., from about 1.50 µg to about 40 µg, from about 2 µg to about 30 µg, from about 3 µg to about 30 µg, from about 4 µg to about 30 µg, from about 5 µg to about 30 µg, from about 10 µg to about 30 µg, from about 10 µg to about 20 µg or from about 15 µg to about 25 µg.

[0049] More specifically, the daily dose of vitamin D3 may be from about 5 µg to about 30 µg, such as, e.g., from about 10 µg to about 20 µg.

[0050] In a further use according to the invention, the strontium component may be administered in a dose corresponding to a daily dose of from about 0.3 g to about 1 g, the alpha-ketoglutarate component may be administered in a dose corresponding to a daily dose of from about 2 g to about 7 g and vitamin D3 may be administered in a dose corresponding to a daily dose of from about 10 µg to about 20 µg.

[0051] Another active form of vitamin D to be used in a method according to the invention is vitamin D2. The daily dose of vitamin D2 may be at least 1 µg, such as, e.g. at least about 1.50 µg, at least about 2 µg, at least about 3 µg, at least about 4 µg, at least about 5 µg, at least about 10 µg, at least about 15 µg, at least about 20 µg, at least about 25 µg, at least about 30 µg, at least about 46 µg, at least about 50 µg, at least about 60 pig, at least about 70 µg, at least about 80 µg, at least about 90 µg, at least about 100 µg, at least about 110 µg, at least about 120 µg or at least about 125 µg or from about 1 µg to about 125 µg such as, e.g., from about 1.50 to about 120 µg, from about 2 µg to about 110 µg, from about 3 µg to about 100 µg, from about 4 µg to about 90 µg, from about 5 µg to about 80 µg, from about 5 µg to about 125 µg, from about 10 µg to about 70 µg, from about 10 µg to about 60 µg, from about 10 µg to about 50 µg, from about 10 µg to about 40 µg, from about 10 µg to about 30 µg, from about 10 µg to about 20 µg, or from about 15 µg to about 25 µg.

[0052] More specifically, the daily dose of vitamin D2 is from about 5 µg to about 125 µg, such as, e.g., from about 10 µg to about 20 µg.

[0053] Other functional equivalents of vitamin D3 and D2, such as alphacalcidol, calcitriol or dihydrotachysterol, may also be administered according to the invention. Alpha-calcidiol, 1α-hydroxy-cholecalciferol, may be administered in amounts of 0.2-3 µg/day, preferably 0.25-2 µg/day. Calcitriol, 1,25-dihydroxy-cholecalciferol, may be administered in amounts of 0.1-10 µg/day, preferably 0.125-2 µg/day and dihydrotachysterol, a vitamin D2 analogue, may be administered in amounts of 0.1-3 mg/day, preferably 0.2-0.6 mg/day.

[0054] In yet a further use, the administration of the strontium component, the alpha-ketoglutarate or amino acid component, if relevant, and vitamin D takes place simultaneously.

[0055] In another use, the administration of the strontium component, the alpha-ketoglutarate or amino acid component, if relevant, and/or vitamin D take place sequentially.

[0056] Calcium is another example of an active substance that may be administered as part of the same treatment as the administration of strontium salt. Calcium is the most abundant mineral in the body, and a major constituent of bone and teeth as calcium phosphate and calcium carbonate. Calcium is also essential in intra- and extracellular fluid exchange, blood clotting, and in maintaining a regular heartbeat. It is also important in the initiation of neuromuscular as well as metabolic functions. Most of the calcium in the body is stored in the bones.

[0057] Thus, calcium is an important participant in many processes in the body, and administration of calcium may have a therapeutic and/or prophylactic effect on many of the diseases and conditions mentioned above.

[0058] Accordingly, the present invention relates to a method as described above further comprising administering a daily dose of calcium.

[0059] In a specific use according to the invention the daily dose of calcium is from about 0.5 g to about 2 g such as, e.g., 0.5 g to about 1.5 g, from 0.5 g to 1 g and from about 1 g to about 1.5 g.

[0060] In yet another use according to the invention, the strontium component is administered in a dose corresponding to a daily dose of from about 0.3 g to about 1 g, the alpha-ketoglutarate or amino acid component is administered in a dose corresponding to a daily dose of from about 2 g to about 7 g and the dose of calcium corresponds to a daily dose of from about 0.5 g to about 1 g.

[0061] The administration of the strontium salt, the alpha-ketoglutarate or amino acid component, if relevant, and calcium may take place simultaneously, either in a single administration form or in separate administration forms for simultaneous administration as described above.

[0062] Alternatively, the strontium salt, the alpha-ketoglutarate or amino acid component, if relevant, and calcium may be administered sequentially.

[0063] Studies have shown that strontium is a full agonist of the calcium-sensing receptor (CaR). Even though the role of the CaR in regulating bone cells is not fully investigated, it appears that strontium and calcium may exert their effect on bone metabolism via the same receptor.

[0064] Accordingly, it may be beneficial not to administer the strontium-containing component and calcium at the same time.

[0065] In one aspect of the present invention, calcium may be administered after the administration of strontium, i.e. the invention relates to a use, wherein calcium is administered at least 0.5 h, such as, e.g., at least 1 h, at least 2 h, at least 3 h, at least 4 h, at least 5 h, at least 6 h, at least 7 h, at least 8 h, at least 9 h, at least 10 h, at least 11 h or at least 12 h after the administration of the strontium component.

[0066] In another aspect calcium may be administered before the administration of strontium, i.e. the invention relates to a use, wherein calcium is administered at least 0.5 h, such as, e.g., at least 1 h, at least 2 h, at least 3 h, at least 4 h, at least 5 h, at least 6 h, at least 7 h, at least 8 h, at least 9 h, at least 10 h, at least 11 h or at least 12 h before the administration of the strontium component.

[0067] In yet another aspect, the strontium salt and the alpha-ketoglutarate or amino acid component, if relevant, may be administered simultaneously and calcium may be administered at least 1 h, such as, e.g., at least 2 h, at least 3 h, at least 4 h, at least 5 h, at least 6 h, at least 7 h, at least 8 h, at least 9 h, at least 10 h, at least 11 h or at least 12 h after the administration of the strontium salt.

[0068] In a further aspect, the strontium salt and the alpha-ketoglutarate or amino acid component, if relevant, may be administered simultaneously and calcium may be administered at least 1 h, such as, e.g., at least 2 h, at least 3 h, at least 4 h, at least 5 h, at least 6 h, at least 7 h, at least 8 h, at least 9 h, at least 10 h, at least 11 h or at least 12 h before the administration of the strontium salt.

[0069] In yet a further aspect, calcium and vitamin D may be administered simultaneously at least 1 h, such as, e.g., at least 2 h, at least 3 h, at least 4 h, at least 5 h, at least 6 h, at least 7 h, at least 8 h, at least 9 h, at least 10 h, at least 11 h or at least 12 h before the simultaneously administration of a strontium salt and vitamin D.

[0070] In another aspect calcium and vitamin D may be administered simultaneously in the morning, and a strontium salt and vitamin D may be administered simultaneously in the evening.

[0071] A further example of an active substance that may be administered as part of the same treatment as the administration of strontium is parathyroid hormone. Parathyroid hormone is composed of 84 amino acid residues and is released in vivo in response to a decrease in the level of extra cellular calcium. Administration of PTH or fragments thereof in a pharmaceutically relevant dose is known to stimulate bone formation, produce a robust increase in bone mineral density and substantially reduce the occurrence of vertebral and non-vertebral fractures. Parathyroid hormone acts directly on the kidney to diminish urinary calcium, and increases bone resorption via an indirect mechanism involving osteoblasts. Parathyroid hormone also increases the activation of vitamin D by stimulating the activity of 1α-hydroxylase enzyme in the kidney, subsequently leading to a better absorption of calcium and, possibly, strontium.

[0072] A commercially available parathyroid hormone containing drug comprises the 34 N-terminal amino acids region of human parathyroid hormone, which is believed to be the biologically active region.

[0073] Accordingly, in a further use according to the invention an amount of parathyroid hormone, or a fragment or analogue thereof, or a parathyroid hormone related peptide, or a fragment or analogue thereof, is administered as part of the same treatment as the administration of strontium salt. In the following the term "PTH" covers parathyroid hormone, fragments, analogues and functional analogues thereof together with parathyroid related hormone and fragments, analogues and functional analogues thereof. PTH may be used as a combined or sequential administration with strontium and, if relevant, alpha-ketoglutarate.

[0074] The daily dose of PTH, when calculated as recombinant human parathyroid hormone (1-34), may be at least 1 µg, such as, e.g. at least about 2 µg, at least about 3 µg, at least about 4 µg, at least about 5 µg, at least about 10 µg, at least about 15 µg, at least about 20 µg, at least about 25 µg, at least about 30 µg, at least about 35 µg, at least about 40 µg, at least about 50 µg, or at least about 60 µg, or from about 1 µg to about 60 µg such as, e.g., from about 2 to about 50 µg, from about 3 µg to about 40 µg, from about 4 µg to about 40 µg, from about 5 µg to about 40 µg, from about 10µg to about 40 µg, from about 10 µg to about 35 µg, from about 10 µg to about 30 µg, from about 10 µg to about 25 µg, from about 10µg to about 20 µg, from about 15 µg to about 40 µg, from about 20 µg to about 40 µg or from about 20 µg to about 30 µg.

[0075] More specifically, the daily dose of PTH, when calculated as recombinant human parathyroid hormone (1-34), may be from about 10 µg to about 40 µg, such as, e.g., from about 10 µg to about 30 µg, from about 10 µg to about 20 µg, from about 20 µg to about 40 µg or from about 20 µg to about 30 µg.

[0076] The medical use according to the invention may comprise administration of a daily dose of bisphosphonate from about 0.1 mg to 60 mg, such as from about 0.2 mg to about 30 mg, from about 0.2 mg to about 20 mg or from about 0.2 mg to about 10 mg.

[0077] In a combination treatment according to the invention in which a strontium containg compound is given in combination with one or more SERM's, the SERM should be used in a dose as determined previously from clinical investigation of the given SERM.

[0078] In the following is given a more detailed description of individual salts according to the invention. Especially, with respect to strontium alpha-ketoglutarate and amino acid salts of strontium, these salts have two active principles, i.e. the strontium part and the alpha-ketoglutarate or amino acid part. Accordingly, these aspects of the invention include individual dose adjustment by addition e.g. of a separate dose of one of the individual components. However, all details with respect to strontium apply also for all the other strontium salts according to the invention.

[0079] Furthermore, the details and particulars described above for strontium salts apply mutatis mutandis to the individual strontium salts, whenever relevant, as well as details and particular described below for the individual strontium salts apply mutatis mutandis to the strontium salts in general, whenever relevant.

Strontium alpha-ketoglutarate salts and compositions



[0080] Bone consists of an organic matrix comprising predominantly collagen type I, and an inorganic phase comprising calcium phosphate and calcium carbonate. The amino acid sequence of collagen type I is remarkably regular with nearly every third residue being glycine. Proline is also present to a much greater extent in collagen than in most other proteins. Moreover, the sequence glycine - proline - 4-hydroxyproline recurs frequently. Alpha-ketoglutarate (AKG) is presumed to be a bone mineral density increasing and bone strength increasing agent for the treatment of osteoporosis and other bone conditions since alpha-ketoglutarate is a precursor of glutamate, which may be converted to proline, an important component of collagen. Alpha-ketoglutarate also participates in the conversion of proline to 4-hydroxyproline. The proline residues in collagen may be hydroxylated at C-4 by prolyl hydroxylase to form 4-hydroxyproline. For this process alpha-ketoglutarate, molecular oxygen and ascorbate are required.

[0081] Furthermore, alpha-ketoglutarate is a very important intermediate in the citric acid cycle. In the citric acid cycle acetyl CoA is completely oxidized to CO2 via interconversions of various carboxylic acids, including AKG. This results in the reduction of NAD and FAD to NADH and FADH2, whose reducing power is then used indirectly in the synthesis of ATP.

[0082] For the treatment and/or prophylaxis of a cartilage and/or bone disease and/or conditions resulting in a dysregulation of cartilage and/or bone metabolism in a mammal, such as, e.g., a human female or male adult, adolescent or a child, such as, e.g., osteoporosis, osteoarthritis, osteopetrosis, osteopenia and Paget's disease, hypercalcemia of malignancy, periodontal disease, hyperparathyroidism, periarticular erosions in rheumatoid arthritis, osteodystrophy, myositis ossificans, Bechterew's disease, malignant hypercalcemia, osteolytic lesions produced by bone metastasis, bone pain due to bone metastasis, bone loss due to sex steroid hormone deficiency, bone abnormalities due to steroid hormone treatment, bone abnormalities caused by cancer therapeutics, osteomalacia, Bechet's disease, hyperostosis, metastatic bone disease, immobilization-induced osteopenia or osteoporosis, or glucocorticoid-induced osteopenia or osteoporosis, osteoporosis pseudoglioma syndrome, idiopathic juvenile osteoporosis, for the improvement of fracture healing after traumatic or atraumatic fracture, and for the maintenance or increase of energy level, for building up or strengthening muscle tissues and for weight gain, the present inventors have found that the use of a strontium containing compound together with an alpha-ketoglutarate-containing compound has prophylactic and/or therapeutic value in that one or more of the following beneficial effects can be obtained:

vii) an improved bioavailability of strontium and/or alpha-ketoglutarate,

viii) an improved absorption of strontium and/or alpha-ketoglutarate,

ix) a reduction of side effects,

x) a flexible dose adjustment of strontium and alpha-ketoglutarate in order to tailor prevention and/or treatment of a specific disease stage,

xi) an additive and possible synergistic effect of strontium and alpha-ketoglutarate,

xii) a possible reduction in daily dosage

xiii) a possible reduction of the number of different pharmaceutical compositions that a patient must use to achieve a therapeutic effect.



[0083] Thus, it is believed that strontium administered together with alpha-ketoglutarate gives a more efficient prevention and/or treatment than either strontium or alpha-ketoglutarate administered alone. This implies that smaller doses of strontium and alpha-ketoglutarate may be used when administered together, as compared to individual administration of the two compounds. The daily dose of alpha-ketoglutarate needed for the treatment and/or prophylaxis of some of the above-mentioned conditions may be rather large, i.e. the subject in the need of treatment will have to take large amounts of alpha-ketoglutarate at a time, or the frequency of intake of the doses may be high, both being of great inconvenience for the subject. The possibility of using smaller doses of strontium and alpha-ketoglutarate would be much more convenient for the subject in the need of treatment.

[0084] Thus, the invention relates to a pharmaceutical composition comprising a therapeutic and/or prophylactic effective amount of one or more first components containing a strontium compound and one or more second components containing an alpha-ketoglutarate compound, together with one or more physiologically acceptable excipients.

[0085] The above-mentioned strontium salts of organic or inorganic acids and salts of alpha-ketoglutaric acid may be in a composition as described above. The salts may be in hydrate, anhydrous, solvate, polymorphous, amorphous, crystalline, microcrystalline or polymeric form. In one embodiment of the invention only non-radioactive isotopes of strontium are used.

[0086] Below follows examples of salts of alpha-ketoglutaric acid, which may be used to adjust the amount of alpha-ketoglutarate in a pharmaceutical composition comprising strontium alpha-ketoglutarate, or in a pharmaceutical composition comprising a strontium-containing compound and an alpha-ketoglutarate compound as described above. The salt may be a salt of an alkali metal, a mixed salt of alkali metals, an alkaline earth metal, or mixtures thereof.

[0087] Specific examples of salts for use according to the invention may be sodium alpha-ketoglutarate, potassium alpha-ketoglutarate, lithium alpha-ketoglutarate, magnesium alpha-ketoglutarate, calcium alpha-ketoglutarate and mixtures thereof.

[0088] The salt may also be a salt of an amine or an amino acid or an ammonium salt or mixtures thereof. The amine may be selected from methyl amine, ethyl amine, propyl amine, isopropyl amine and butyl amine, and the amino acid may be selected from arginine, ornithine, lysine and histidine.

[0089] As exemplified by the salts mentioned above, the counter ions in the strontium and alpha-ketoglutarate-containing compounds may be active substances having the same medical indications as strontium and alpha-ketoglutarate. Examples of such compounds are e.g., strontium glutamate and calcium alpha-ketoglutarate. A composition containing strontium and alpha-ketoglutarate could therefore contain strontium glutamate and calcium alpha-ketoglutarate. However, treatment of some conditions will require relatively large doses of strontium and alpha-ketoglutarate to be administered. This makes the total amount of such a pharmaceutical composition that has to be administered to the subject in the need of treatment relatively large, which may be of great inconvenience to the subject.

[0090] Described is a strontium salt of alpha-ketoglutaric acid with the following formula I:

        -OOC-CH2-CH2-C(=O)-COO- Sr2+     (I)

which may be in a hydrate, anhydrous, solvate, polymorphous, amorphous, crystalline, microcrystalline or polymeric form.

[0091] The salt of the above-mentioned formula is composed of two active substances, i.e. a bone anti-resorptive and bone anabolic agent in the form of strontium, and then a further amount of a bone mineral density increasing, bone strength and bone quality increasing agent in the form of alpha-ketoglutarate. As compared to previous known strontium salts, such as, e.g., strontium ranelate or strontium chloride, wherein only the strontium ion has a therapeutic and/or prophylactic effect on bone and/or cartilage diseases, both components in the novel salt are active components having a therapeutic and/or prophylactic effect.

[0092] By using the salt in pharmaceutical formulations, it may be possible to reduce the size of the formulations, even though the same dose of strontium and alpha-ketoglutarate is administered as in formulations comprising strontium and alpha-ketoglutarate as separate salts, together with their respective counter ions.

[0093] Furthermore, as described above the combination of strontium and alpha-ketoglutarate in the salt may have additive or synergistic beneficial effects on bone and/or cartilage tissue. Furthermore, the novel salt has suitable properties with respect to physio-chemical properties such as, e.g., water solubility; and it has technical properties suitable for processing the novel salt into pharmaceutical compositions.

[0094] Also disclosed is a process for preparing the novel strontium alpha-ketoglutarate substance according to the invention. The salt may be prepared by any method known to a person skilled in the art for preparing such salt. One example of such a process comprises reacting a salt of strontium with alpha-ketoglutaric acid or a salt thereof, wherein the molar ratio of strontium to alpha-ketoglutaric acid may be 1:1. A more specific example comprises reacting alpha-ketoglutaric acid with strontium hydroxide and/or strontium oxide, wherein the molar ratio of strontium to alpha-ketoglutarate optionally may be 1:1.Yet another example of such a process comprises reacting strontium metal with alpha-ketoglutaric acid. As disclosed in examples 4-7 the invention also relates to specific methods of producing the strontium salt of the invention involving synthesis at temperatures above 100 °C.

[0095] Also disclosed is a method for the treatment and/or prophylaxis of a cartilage and/or bone disease and/or conditions resulting in a dysregulation of cartilage and/or bone metabolism in a mammal, such as the ones mentioned above, for the maintenance or increase of energy level, for building up or strengthening muscle tissues and for weight gain, the method comprising administering to a subject in need thereof an amount of the strontium alpha-ketoglutarate salt according to the invention.

[0096] In the latter method wherein strontium alpha-ketoglutarate is administered, the salt may be administered in a dose corresponding to from about 0.1 to about 17 g daily calculated as anhydrous salt. More specifically, the salt may be administered in a dose corresponding to from about 0.2 to about 15 g daily such as, e.g., from about 0.4 to about 13 g daily, from about 0.6 to about 12 g daily or from about 0.7 to about 11.5 g daily calculated as anhydrous salt.

[0097] As described above, depending on the condition the subject is having, there might be a need for increasing and/or adjusting the amounts of strontium and alpha-ketoglutarate administered. Thus, the invention further relates to a use, which comprises the administration of an additional dose of a strontium-containing compound and/or an additional dose of an alpha-ketoglutarate-containing compound together with strontium alpha-ketoglutarate. The strontium-containing and alpha-ketoglutarate containing compounds may be selected from the compounds described above.

[0098] For the use wherein the strontium alpha-ketoglutarate salt is administered, optionally together with an additional amount of strontium and/or alpha-ketoglutarate, and the method wherein one or more strontium-containing compounds and one or more alpha-ketoglutarate-containing compounds are administered, the weight ratio between the total daily dose of strontium and the total daily dose of alpha-ketoglutarate may be from about 0.01 to about 4, such as, e.g., from about 0.01 to about 4, from about 0.01 to about 2, from about 0.01 to about 1, from about 0.01 to about 0.6, from about 0.03 to about 4, from about 0.03 to about 2, from about 0.03 to about 1, from about 0.1 to about 2, from about 0.1 to about 1, from about 0.15 to about 0.5, about 0.2 to about 2, from about 0.3 to about 2 or from about 0.4 to about 2.

[0099] The daily dose of alpha-ketoglutarate may be at least about 0.5 g, such as, e.g., at least about 1.0 g, at least about 1.5 g, at least about 2.0 g, at least about 2.5 g, at least about 3.0 g, at least about 4 g, at least about 5 g or from about 0.5 g to about 10 g, from about 0.5 g to 7 g, from about 2 g to about 10 g or from about 2 g to about 7 g.

[0100] In a use according to the invention, the strontium component and the alpha-ketoglutarate component may be administered in single compositions or simultaneously in separate, co-administered compositions. Strontium and alpha-ketoglutarate may be in the form of strontium alpha-ketoglutarate, optionally with an additional amount of a strontium-containing compound and/or an alpha-ketoglutarate-containing compound. The additional component may be added to the strontium alpha-ketoglutarate in the same composition, or may be in a separate composition intended for simultaneous administration.

[0101] Strontium and alpha-ketoglutarate may also be combined as a mixture of one or more strontium-containing compounds and one or more alpha-ketoglutarate-containing compounds in the same formulation, or in separate forms intended for simultaneous administration. When two or more separate formulations are being co-administered, each formulation, especially those for use by oral route, may be color-coded or otherwise easily identifiably labeled in order to avoid confusion by the subject or physician.

[0102] In another use according to the invention, the administration of the strontium component and the alpha-ketoglutarate component may take place sequentially.

Strontiumglutamate compositions.



[0103] As mentioned above alpha-ketoglutarate may be converted to the amino acid glutamate, which is a precursor of glutamine, arginine and proline, the latter being an important component of collagen. Thus, the amino acid glutamate is also considered an important agent in the treatment of cartilage and/or bone condition, and the administration of strontium and glutamate together in the form of a strontium glutamate salt is believed to have prophylactic and/or therapeutic value in that one or more of the beneficial effects mentioned above for strontium and alpha-ketoglutarate can be obtained. In addition glutamate may directly affect specific glutamate receptors present on the resorbing osteoclasts, and thus affect the metabolic activity and bone resorbing action of these cells.

[0104] Thus, also disclosed is the use of a strontium glutamate salt of the formula II:

        (-OOC-C(NH3+)H-CH2-CH2-COO-)2 Sr2+     (II)

as a medicine as well as to a method for the preparation of strontium glutamate. The invention also relates to specific methods of producing the strontium salt of the invention involving synthesis at temperatures above 100 °C.

[0105] Also diclosed is the use of strontium glutamate for the preparation of a pharmaceutical composition for the treatment and/or prophylaxis of a cartilage and/or bone disease and/or conditions resulting in a dysregulation of cartilage and/or bone metabolism in a mammal, such as, e.g., a human female or male adult, adolescent or a child, such as described herein before.

[0106] For the treatment and/or prophylaxis of different cartilage and/or bone diseases, the possibility of administering various amounts of strontium and glutamate respectively may be desired. The amount of strontium and glutamate in a pharmaceutical composition according to the invention may be adjusted by adding an additional amount of strontium in the form of a strontium-containing compound and/or an additional amount of glutamate in the form of a glutamate-containing compound to the composition.

[0107] All the strontium salts of organic or inorganic acids mentioned above and salts of glutamic acid may be used to adjust the amount of strontium and glutamate in a pharmaceutical composition comprising strontium glutamate, and may also be used in a composition according to the invention comprising a strontium-containing compound and a glutamate-containing as described above. The salts may be in hydrate, anhydrous, solvate, polymorphous, amorphous, crystalline, microcrystalline or polymeric form.

[0108] The organic acids for making strontium salts may be selected from any of the groups described above.

[0109] Below follows examples of salts of glutamic and aspartic acid, which may be used to adjust the amount of glutamate and/or aspartate compound in a pharmaceutical composition comprising strontium glutamate and/or strontium aspartate, or in a pharmaceutical composition comprising a strontium-containing compound and a glutamate compound as described above.

[0110] The salt may be a salt of an alkali metal, a mixed salt of alkali metals, an alkaline earth metal, or mixtures thereof.

[0111] Specific examples of salts for use according to the invention may be sodium glutamate, potassium glutamate, lithium glutamate, magnesium glutamate, calcium glutamate. The salt may also be a salt of an amine or an amino acid or an ammonium salt or mixtures thereof. The amine may be selected from methyl amine, ethyl amine, propyl amine, isopropyl amine and butyl amine, and the amino acid may be selected from arginine, ornithine, lysine and histidine.

[0112] Also disclosed is a method for the treatment and/or prophylaxis of a cartilage and/or bone disease and/or conditions resulting in a dysregulation of cartilage and/or bone metabolism in a mammal, such as, e.g., a human female or male adult, adolescent or a child, such as the ones mentioned above, for the maintenance or increase of energy level, for building up or strengthening muscle tissues and for weight gain, the method comprising administering to a subject in need thereof an amount of the strontium glutamate salt according to the invention.

[0113] In the latter method wherein strontium glutamate is administered, the salt may be administered in a dose corresponding to from about 0.2 to about 28 g daily calculated as anhydrous salt. More specifically, the salt may be administered in a dose corresponding about 0.3 to about 25 g daily such as, e.g., from about 0.7 to about 20 g daily, from about 1 to about 17 g daily or from about 1.2 to about 16 g or from about 2 to about 6 g daily calculated as anhydrous salt.

[0114] As described above, depending on the condition the subject is having, there might be a need for increasing and/or adjusting the amounts of strontium and glutamate. Thus, further embodiments of the invention comprise the administration of an additional dose of a strontium-containing compound together with strontium glutamate or the administration of an additional dose of a glutamate-containing compound. The strontium-containing and glutamate-containing compounds may be selected from the compounds described above.

[0115] For the use wherein a strontium glutamate salt is administered, optionally together with an additional amount of strontium and/or L- or D-glutamate, and the use wherein one or more strontium-containing compounds and one or more L- or D-glutamate-containing compounds are administered, the weight ratio between the total daily dose of strontium and the total daily dose of glutamate is from about 0.01 to about 4, such as, e.g., from about 0.01 to about 4, from about 0.01 to about 2, from about 0.01 to about 1, from about 0.01 to about 0.6, from about 0.03 to about 4, from about 0.03 to about 2, from about 0.03 to about 1, from about 0.1 to about 2, from about 0.1 to about 1, from about 0.15 to about 0.5, from about 0.2 to about 2, from about 0.3 to about 2 or from about 0.4 to about 2.

[0116] As mentioned above, the daily dose of strontium may be at least about 0.01 g, such as, e.g., at least about 0.025 g, at least about 0.050 g, at least about 0.075 g, at least about 0.1 g, at least about 0.2 g, at least about 0.3 g, at least about 0.4 g or at least about 0.5 g or from about 0.01 g to about 2 g such as, e.g., from about 0.1 g to about 2 g, from about 0.3 g to about 2 g or from about 0.3 g to about 1 g.

[0117] The daily dose of L- and/or D-glutamate may be at least about 0.5 g, such as, e.g., at least about 1.0 g, at least about 1.5 g, at least about 2.0 g, at least about 2.5 g, at least about 3.0 g, at least about 4 g, at least about 5 g or from about 0.5 g to about 10 g, from about 0.5 g to 7 g, from about 2 g to about 10 g or from about 2 g to about 7 g.

[0118] The invention also relates to a use wherein the strontium component and the glutamate component are administered in the form of a pharmaceutical composition as described above.

[0119] In a use according to the invention the strontium component and the glutamate component may be administered in single mixture formulations or simultaneously in separate, co-administered formulations. Strontium and glutamate may be in the form of strontium glutamate, optionally with an additional amount of a strontium-containing compound and/or a glutamate-containing compound. The additional component may be added to the strontium glutamate in the same formulation, or may be in a separate formulation intended for simultaneous or sequential administration.

[0120] Strontium and glutamate may also be combined as a mixture of one or more strontium-containing compounds and one or more glutamate-containing compounds in the same formulation, or in separate forms intended for simultaneous administration. When two or more separate formulations are being co-administered, each formulation, especially those for use by oral route, may be color-coded or otherwise easily identifiably labeled in order to avoid confusion by the subject or physician.

[0121] As described above one or more active substances may be added to a pharmaceutical composition according to the invention or administered as part of the same treatment as the administration of strontium and glutamate.

Strontiumaspartate compositions



[0122] Aspartate is an amino acid structurally related to glutamate, which can also form pharmaceutically relevant salts in complex with strontium. As all amino acids except glycine aspartate exist in an L-form, which is the physiologically relevant form used in all biological systems and the 'mirror image' enantiomer denoted D-Aspartate. D-aspartate may directly or indirectly affect metabolism of bone and/or cartilage via binding to the N-methyl-D-aspartate (NMDA) receptor, which has been found on metabolically active osteoclasts and may also be present on chondrocytes of articular cartilage. Thus the present invention also relates to a strontium aspartate salt (in D- or L- form or a mixture thereof) of the formula (III)

        (-OOC-C(NH3+)H-CH2-COO-)2 Sr2+     (III)

as a medicine as well as to a method for the preparation of strontium aspartate. Also disclosed are specific methods of producing the strontium salt of the invention involving synthesis at temperatures above 100 °C.

[0123] Also disclosed is the use of strontium aspartate for the preparation of a pharmaceutical composition for the treatment and/or prophylaxis of a cartilage and/or bone disease and/or conditions resulting in a dysregulation of cartilage and/or bone metabolism in a mammal, such as, e.g., a human female or male adult, adolescent or a child, such as, e.g., male and female osteoporosis, osteoarthritis, osteopetrosis, osteopenia and Paget's disease, hypercalcemia of malignancy, periodontal disease, hyperparathyroidism, periarticular erosions in rheumatoid arthritis, osteodystrophy, myositis ossificans, Bechterew's disease, malignant hypercalcemia, osteolytic lesions produced by bone metastasis, bone pain due to bone metastasis, bone loss due to sex steroid hormone deficiency, bone abnormalities due to steroid hormone treatment, bone abnormalities caused by cancer therapeutics, osteomalacia, Bechet's disease, hyperostosis, metastatic bone disease, immobilization-induced osteopenia or osteoporosis, or glucocorticoid-induced osteopenia or osteoporosis, osteoporosis pseudoglioma syndrome, idiopathic juvenile osteoporosis, for the improvement of fracture healing after traumatic or atraumatic fracture, and for the maintenance or increase of energy level, for building up or strengthening muscle tissues and for weight gain. The pharmaceutical composition may further comprise one or more physiologically acceptable excipients.

Pharmaceutical compositions



[0124] The pharmaceutical compositions used in the invention normally comprise the specific compounds together with one or more physiologically acceptable excipients, i.e. a therapeutically inert substance or carrier.

[0125] The carrier may take a wide variety of forms depending on the desired dosage form and administration route.

[0126] The pharmaceutically acceptable excipients may be e.g. fillers, binders, disintegrants, diluents, glidants, solvents, emulsifying agents, suspending agents, stabilizers, enhancers, flavors, colors, pH adjusting agents, retarding agents, wetting agents, surface active agents, preservatives, antioxidants etc. Details can be found in pharmaceutical handbooks such as, e.g., Remington's Pharmaceutical Science or Pharmaceutical Excipient Handbook.

[0127] Above are mentioned specific examples of the amounts of compounds administered. However, it will be understood that the amount of the compounds actually administered will be determined by a physician in light of the relevant circumstances including the condition to be treated, the choice of compounds to be administered, the age, weight, and response of the individual patient, the severity of the patient's symptoms and the chosen route of administration. While the present compounds are preferably administered orally, the compounds may also be administered by any other suitable route.

[0128] The pharmaceutical composition comprising a compound according to the invention may be in the form of a solid, semi-solid or fluid composition.

[0129] The solid composition may be in the form of tablets such as, e.g. conventional tablets, effervescent tablets, coated tablets, melt tablets or sublingual tablets, pellets, powders, granules, granulates, particulate material, solid dispersions or solid solutions.

[0130] In one embodiment of the invention, the pharmaceutical composition may be in the form of a tablet. The tablet may be coated with a coating that enables release of at least part of the salt in the proximal part of the small intestine, such as e.g. the duodenum and/or the proximal jejunum, such as at least 50% w/w, at least 60% w/w, at least 65% w/w, at least 70% w/w, at least 80% w/w or at least 90% w/w of the total amount of the salt contained in the tablet.

[0131] The tablet may have a shape that makes it easy and convenient for a patient to swallow. The tablet may thus e.g. have a rounded or a rod-like shape without any sharp edges. Furthermore, the tablet may be designed to be divided in two or more parts.

[0132] A semi-solid form of the composition may be a paste, a gel or a hydrogel.

[0133] The fluid form of the composition may be a solution, an emulsion including nano-emulsions, a suspension, a dispersion, a liposomal composition, a spray, a mixture, a syrup or an elixir.

[0134] Other suitable dosages forms of the pharmaceutical compositions according to the invention may be capsules, sachets, troches, devices etc.
The pharmaceutical compositions may be prepared by any of the methods well known to a person skilled in pharmaceutical formulation.

[0135] Also disclosed is that the use of a composition according to the invention may lead to improved fracture healing after traumatic or atraumatic fracture, where the fracture e.g. may be one of the following traumatic or atraumatic fractures: fracture to the distal radius, such as e.g. a Colle's fracture or a Smiths fracture, a fracture of the femur, such as e.g. the proximal femur, such as e.g. a cervical fracture, a trochanteric fracture or a subtrochanteric fracture.

[0136] The improved fracture healing may be defined in terms of a reduction of the time a patient will require a plaster, reduction of the time to healing as defined on a X-ray, reduction in the time to fracture stability, improvement of callus formation as viewed by X-ray, reduction in time before appearance of callus formation as viewed by X-ray and/or reduction in time for regaining full or near-full mobility or physical activity level.

[0137] Other aspects of the invention

[0138] Other embodiments of the invention appear from the appended claims. The details and particulars described above and relating to the compounds and compositions according to the invention apply mutates mutandis to the other aspects of the invention.

Legends to figures



[0139] 

Figure 1: Molecular structure of strontium L-glutamate (6*H2O) in the crystalline form as disclosed by Schmidbaur et al 1989. The crystal is shown with atoms depicted as ellipsoids (defined at the 50% probability level according to the ORTEP program (Oak Ridge Thermal Ellipsoid Plot Program)). Complexation by Sr2+ by three additional symmetrically related amino acids is indicated only by the coordinating carboxy oxygen atoms O1, O2 and O3/O4.

Figure 2: Extended layer structure of strontium L-glutamate (6*H2O) in the crystalline form as disclosed by Schmidbaur et al 1989. Strontium atoms are shown as gray and interstitial water molecules as black.

Figure 3: Molecular structure of strontium L-aspartate (3*H2O) in the crystalline form as disclosed by Schmidbaur et al 1990. The crystal is shown with atoms depicted with arbitrary radii. Complexation by Sr2+ by four additional symmetrically related amino acids is indicated only by the coordinating carboxy oxygen atoms O3'/O3" and O4"/O4"'. Complexation to oxygen atoms of two water molecules are visible as 06 and 07.

Figure 4: Diffractograms of the x-ray analysis of two strontium salts. The top diffractogram shows Strontium glutamate hexahydrate, as synthesised by strontium hydroxide and L-glutamic acid at high temperature but using the raction conditions described in example 2.

Figure 5. X-ray diffractogram of crystals of strontium glutamate hexahydrate prepared by the method as described in example 7.

Figure 6 (Reference). X-ray diffractogram of crystals of strontium malonate prepared by the method as described in example 7. The malonate salt of strontium have not previously been characterized and comprise a new crystallographic structure, but it is apparent from the stable baseline, and well defined spacing of diffraction peaks, that the crystal form of the malonate salt is homogeneous and pure.

Figure 7. Results of the optimisation experiments for strontium glutamate synthesis outlined in table 9. The influence on the yield of the synthesis of strontium glutamate was investigated by varying four parameters. (Yields above 100% indicate incomplete drying).

Figure 8 (previous page): Plot of serum strontium concentrations measured in rats given a single dose of strontium as indicated in the upper part of each panel. The data points represent mean and standard deviation for each measuring point. Pre-dosis represent corresponding samples taken from animals treated with vehicle alone.

Figure 9. Modelling of theory (solid line) to experimental data (diamonds) of strontium with error bars superimposed. The theory fits the data excellently and the area under the curves (AUC) are calculated by the model. The strontium content always peaked after 60 min. but the second peak defined by the rate of metabolism vary between the salts. In the present examples, the Sr-α-ketoglutrate is quickly metabolised while Sr-glutamate rests longer in the serum.


Examples


Example 1 (Reference)


General method for preparation of crystalline salts of strontium by precipitation from dissolved strontium chloride and dissolved sodium salts of the appropriate carboxylic anions



[0140] In a glass-beaker of 100 mL volume, 5 g of the sodium salt of the carboxylic acid was dissolved in a small volume of water that was slightly heated at temperatures not greater than 30-50 °C. The final volume was 25-50 mL. In another beaker 10 g of SrCl2 (SrCl2 hexahydrate, Sigma-Aldrich 43,966-5) was dissolved in 100 mL of water. This latter solution was slowly decanted into the first solution of the dissolved sodium salt. The transfer continued until an initial cloudiness was observed, which resulted in a total volume of 50-100 mL. The solution was allowed to rest at room temperature (22-24 °C) for several days until significant amounts of crystallized precipitate of the organic strontium salt appeared.

[0141] The reaction that proceeds is exemplified by the reaction between strontium ions and sodium fumarate (reaction schemes (a) and (b)):

        NaOOCCHCHCOONa(s)+H2O(l)→-OOCCHCHCOOH(aq)+2Na+(aq)+OH-(aq)     (a)

        -OOCCHCHCOOH(aq)+Sr2+(aq)→Sr(OOCCHCHCOO)(aq)+H+(aq)     (b)



[0142] In order to accelerate the crystallisation, we have found that addition of small volumes of ethanol, such as from 5 -10 vol/vol % to 50 - 60 % vol/vol induces a significant acceleration of the precipitation of the desired strontium salt. Addition of ethanol is of special importance in the synthesis of strontium salts with solubility exceeding 2 g/l at room temperature (22-24 °C), and will thus provide a substantial benefit for the synthesis of strontium salts of L-aspartate, L-glutamate and lactate. In order to reach the required product within a short period, it was essential to observe an initial crystallisation or an initial dimness in the solution right from the first stage.

[0143] After the precipitation, the solution was filtered on a Büchner funnel using a suction flask and the crystals were flushed in small volumes of ethanol. Crystals of some of the salts were very soluble, so in order to improve the yield of crystals, the solution was allowed to rest longer, such as at least 30 - 60 min. Repeated crystallisation resulted in yields of approx. 50%. Strontium salts of L-aspartate and of lactate were very soluble, with solubility exceeding 25 g/l in water at room temperature.

[0144] The lactate and L-glutamate salts of strontium were precipitated from solutions with an excess of strontium chloride and large crystals of the lactate salt were achieved by slow evaporation of the solvent.

Example 2 (Reference)


General method for preparation of crystalline salts by neutralisation of carboxylic acids with strontium hydroxide



[0145] A small amount of the organic acid proper (0.75 - 3 g, see table below) was dissolved in water by heating to temperatures between 30°C - 50 °C. Then, strontium hydroxide (Sigma Aldrich, Sr(OH)2*8H2O, MW 265.71, CAS no. 1311-10-0, approx. 10 g/L) was slowly added. Then, a magnetic stirring rod was added and the stirring and gentle heating (i.e. 30 - 50°C) of the suspension was started. After some time, the solution clarifies and all the solid material dissolves. The heating is maintained, and after three hours of incubation, the solution is filtered while hot on a Büchner funnel. Very small amounts of impurities were left in the filter.

[0146] The filtrate was subsequently allowed to cool at room temperature overnight, which resulted in growth of fine-powdered crystals of the desired strontium salt. Further purifications of the salts can be performed by repeated re-crystallizations (table 6).
Table 6: Amounts of start reagent used for organic strontium salt synthesis and recoveries in the synthesis of eight specific organic strontium salts following the general reaction pathway with free-acid forms of the anion, and strontium hydroxide
Strontium salt of (free acid used): Sr(OH)2 *8H2O Free acid Amount obtained Recovery* Melting Temp. Solubility structure
Fumarate1 2.044 g 1.140 g 0.999 g 99 % >380°C Yes No
α-ketoglutarate2 2.017 g 1.441 g 0.828 g 72 % >380°C. Yes No
succinate 2.098 g 1.177 g 0.958 g 92 % 230°C Yes Yes
L-Ascorbate3 2.094 g 1.805 g 2.005 g 15 % >380°C Yes No
L-Glutamate 2.017 g 1.453 g 0.175 g 15 % >380°C Yes Yes
Citrate 2.057 g 1.918 g 1.123 g 48 % >380°C Yes Yes
D-Aspartate 2.190 g 1.316 g 0.167 g 14 % >380°C No No
Tartrate 2.070 g 1.502 g 2.005 g 129 % >380°C. Yes Yes
Notes
*) Recovery calculated in % of the strontium content in Sr(OH)2 *8H2O.
1) Fumaric acid is insoluble in water, and ethanol is added to the suspension until complete solubilization is achieved. The synthesis is continued with this material.
2) The strontium-AKG salts has a slight brownish appearance and a melting temperature %
3) In addition to the indicated amounts of strontium hydroxides and L-ascorbate an additional 4.087g SrCl2*6H2O solubilized in water is added to the reaction mixture.

Example 3


Determinations of solubility of organic strontium salts


Synthesis of strontium salts



[0147] The great majority of strontium salts could be obtained by reacting the sodium salt of the organic acid with strontium chloride following the general synthesis method described in example A. However, strontium citrate, strontium tartrate, strontium succinate and strontium α-ketoglutarate for the solubility investigations was obtained by synthesis from the free acid forms of the carboxylic acid and strontium hydroxide as described in example 2. Strontium glutamate was obtained as described in example 4, using an incubation temperature of 100°C and using strontium chloride and L-glutamic acid for the synthesis for obtaining pure and homogeneous hexahydrate crystals of strontium glutamate. As described in example 4 the strontium glutamate salt obtained by this method is distinct from a previously described form of crystalline strontium L-glutamate. Detailed investigations of solubility were carried with the strontium salts listed in table 7:
Table 2: Overview of strontium salts used in investigation of solubility. MW indicates the molecular weight of the homogeneous crystalline form of the salt with the indicated amount of crystal water and % Sr gives the molar percentage that strontium constitutes of this crystalline form
Strontium salt MW % Sr
Sr-ranelate (*7H2O) * 639.6 27.4
SrCl2 (*6H2O) * 266.6 32.9
Sr-fumarate (*6H2O) * 309.7 28.3
Sr-L-glutamate (*6H2O) 340.7 25.7
Sr-α-ketoglutarate (*6H2O) 339.7 25.8
Sr-aspartate (*3H2O) 272.7 32.1
Sr-succinate (*6H2O) 311.7 28.1
Sr-ascorbate (*6H2O) * 545.8 16.1
Sr-malelate (*6H2O) 309.7 28.3
Sr-malonate (*1H2O) * 207.7 42.2
Sr-pyruvate (*6H2O) 369.7 23.7
Sr-tartrate (*6H2O) * 343.7 25.5
Sr-citrate (*6H2O) * 749.1 35.1
Reference compounds


[0148] The solubility of the organic carboxylic acid strontium salts, were measured in water. The solubility of these salts was also measured as a function of temperature. This was performed by incubating the saturated solutions of the salts in temperature-controlled incubators. Furthermore the solubility of the salts was studied in pure distilled water as well as a 0.05 M ammonium carbonate buffered solutions, with a physiological pH of 7.5.

[0149] The buffered solutions were immersed into a bath of water temperature controlled at either room temperature (22 - 24 °C), at 30 °C or at 40 °C. The test tubes were stirred and the solutions were subsequently incubated in an incubater with constant temperature for 24 hours. In order to eliminate any reminiscent strontium chloride influence on the determination of solubility, all the precipitate was collected at the bottom of the test tubes and the solutions above the precipitate were carefully removed and substituted by fresh solutions. After substitution of the solutions, the test tubes were stirred again and allowed to rest for another 24 hours. From these solutions, the dissolved proportions of the strontium salt were collected in volumes of 1 mL at the specified temperature. The solutions were diluted to 50 mL before analysis by Flame Atomic Absorption Spectrometry (F-AAS). Before subsequent series of sampling, the solutions were equilibrated at the next temperature for 24 hours.

Analysis of Strontium by flame atomic absorption spectrometry F-AAS



[0150] Two methods were used for quantification of strontium in solutions: Flame Atomic Absorption Spectrometry (F-AAS), and the more sensitive inductively-coupled-plasma-mass spectrometry (ICP-MS). For most investigations, the F-AAS method had sufficient sensitivity.

[0151] Some of the very soluble strontium salts were further diluted before analysis by F-AAS. The measurements were performed by using a Perkin-Elmer 2100 equipped with a hydrogen lamp for correction of the background signal. Strontium was measured at a slit with of 0.2 nm, the wavelength was 460.8 nm operated at an energy of 58 and a current of 8 mA.

Temperature and pH influence on organic strontium salt solubility



[0152] For the majority of the organic strontium salts listed in table 2, temperature changes in the interval from 20 - 40 °C had only little influence on solubility (table 3). However, for strontium L-glutamate a significant influence of temperature on solubility was observed in the range between 20 °C and 40 °C. The solubility of this salt increased more than threefold in the investigated interval in contrast to most other salts. It is noted, that the solubility under physiological conditions (37 °C), is of relevance for the pharmaceutical use of the substances, and thus the surprising increase in strontium glutamate solubility at higher temperature may have great potential therapeutic implications.

[0153] The solubility of the strontium salts in an ammonium carbonate buffered solution of pH 7.5 was generally higher than the solubility determined in pure water (table 8). However, there were some notable exceptions, such as strontium maleate, which had decreased solubility in the buffered solution. Accordingly, it was found most relevant to compare the solubility of the strontium salts by comparing the values obtained in water, as shown in table 8.

Relative solubility



[0154] The water-solubilities of the organic strontium salts at room temperature and at 40 °C, are listed in table 8. The strontium salts of L-aspartate and of lactate had solubilities exceeding 50 g/l hampering exact determination of solubility with the employed experimental procedures.

[0155] The results correspond to the observations during the synthesis experiments where the citrate, the fumerate and the tartrate precipitated instantly when synthesized by the production procedures described in examples 1 and 2. This is indicative of a poor solubility of these strontium salts, as apparent by the lower solubility of these salts compared to the other organic strontium salts at both 22 °C and 40 °C.

[0156] The glutamate salt showed a higher solubility than the other salts, especially at a temperature of 40 °C. During the synthesis of this salt, it was necessary to add alcohol to the solution, to initiate crystal growth, indicative of relatively high water solubility. The other studied strontium salts only precipitated after evaporation of the solvent for a few days at room temperature, but addition of alcohol was not required to initiate crystal formation and precipitation.
Table 8. Relative solubility in water buffered solutions at pH 7.5 at 40°C and room temperature (22 - 24°C) of the investigated Strontium-salts, as determined by F-AAS.
STRONTIUM SALT SOLUBILITY AT ROOM TEMPERATURE (22 - 24°C) (mg/L) SOLUBILITY AT 40°C (mg/L)
Anion In water pH 7.5 In water pH 7.5
Malonate** + 1474 2816 1441 2127
L-glutamate** 2111 3022 7093 7195
L-aspartate** 4200   7900  
Pyruvate* 2204 1946 1929 1829
α-ketogluterate** 1316 2252 3534 3809
Fumerate** + 571 1215 444 977
Maleate** 3002 1680 2527 1457
Tartrate** + 883 1831 1028 1400
Ranelate**** + 760 890 1450 1970
Succinate** 1137 926 1116 2233
Citrate*** + 107 388 147 430
*) Mono-carboxylic acid
**) Di-carboxylic acid
***) Tri-carboxylic acid
****) Quattro-carboxylic acid
+ Reference compounds

Example 4


Preparation of strontium glutamate hexahydrate by synthesis at 100°C



[0157] Initially, a suspension of glutamic acid (white colored) is prepared by adding 100 mL of millipore water to 14.703 g (0.1 moles) of solid L-glutamic acid (Sigma Aldrich, C5H9NO4, MW 187.14 g/mole, CAS no. 142-47-2, lot. no. 426560/1, filling code 43003336) in a 250 mL beaker. To this suspension was added 26.66 g (0.1 moles) of solid SrCl2 (SrCl2 hexahydrate, Sigma-Aldrich 43,966-5, MW 266.6). Then, a magnetic stirring rod was added and the stirring and heating was started to the point of boiling of the suspension. The final suspension is also white colored and the stirring is sustained by maintaining a medium rotation rate of the stirring apparatus. In order to prevent carbon dioxide from entering the solution, the beaker was covered by a covering glass.

[0158] After some minutes of boiling and stirring, the solution clarified and all the solid material dissolved. The boiling was maintained, and additional water was added when required, as to replace the water lost by boiling. After three hours of boiling, the solution was filtered while boiling on a Büchner funnel. Very small amounts of impurities were left in the filter. The filtrate was subsequently allowed to cool to room temperature, which resulted in growth of fine-powdered crystals of strontium glutamate hexahydrate. Precipitation of the final product progressed in the filtrate within an hour. The product was filtered and dried at 110°C in an oven for ½ hour followed by drying 12 hours In a dessicator over silica orange. Before analysis by x-ray crystallography and by FAAS, the salts were ground to fine powder by a mortar.

[0159] The X-ray crystalographic analysis (figure 4) revealed that the synthesized strontium glutamate salt was distinct from the previously described strontium L-glutamate hexahydrate salt described in figures 1 and 2 and tables 2 and 3.

[0160] This salt and the resulting diffractogram corresponds to the strontium L-glutamate hexahydrate salt previously described (H. Schmidbaur, I. Bach, L. Wilkinson & G. Müller (1989), Chem Ber. 122; 1433-1438) and further detailed in figure 1 and 2 and table 2 and 3. The lower trace shows a strontium glutamate hexahydrate salt synthesized from strontium chloride and L-glutamic acid as disclosed in the present example.

[0161] The total yield of strontium glutamate hexahydrate was approximately 92% before recrystallisation, and the majority of impurities consisted of reminisces of the reagents and of strontium carbonate. This yield is significantly higher than the yield obtained by synthesis under conventional conditions where only 15 % was obtained (please see example 2). Thus, the high temperature synthesis method as disclosed in this patent provides a significant gain in yield and a reduction in synthesis time, while resulting in a strontium glutamate salt of higher purity. Furthermore, the strontium glutamate obtained by this synthesis procedure was distinct from the strontium L-glutamate hexahydrate salt previously described (H. Schmidbaur, I. Bach, L. Wilkinson & G. Müller (1989), Chem Ber. 122; 1433-1438). The strontium glutamate hexahydrate described previously in the literature by Schmidbaur et al was reported to have very low solubility (0.023 g/l), whereas the strontium glutamate salt prepared by the method disclosed in the present example had a solubility above 2 g/l. This later parameter is very important for potential medical use of the strontium salt as described in the present invention.

[0162] Further improvements of the synthesis may include degassing by nitrogen or by argon of the water and of all aqueous solutions, which prevents contact to carbon dioxide that eventually may lead to formation of impurities of strontium carbonate. It follows that a person skilled in the art will easily be able to adapt the procedure to proceed under an inert gas atmosphere.

Example 5


Preparation of strontium aspartate trihydrate by synthesis at 100 °C



[0163] Initially, a suspension of aspartic acid (white colored) is prepared by adding 100 mL of millipore water to 13.311 g (0.1 moles) of solid L-aspartic acid (Fluka, C5H9NO4, MW 133.11 g/mole, CAS no. 56-84-8, lot. no. 432866/1, filling code 52603495) in a 250 mL beaker. To this suspension was added 26.571 g (0.1 moles) of solid strontium hydroxide (Sigma Aldrich, Sr(OH)2*8H2O, MW 265.71, CAS no. 1311-10-0). Then, a magnetic stirring rod was added and the stirring and heating was started to the point of boiling of the suspension. The final suspension is also white colored and the stirring is sustained by maintaining a medium rotation rate of the stirring apparatus. In order to prevent carbon dioxide from entering the solution, the beaker was covered by a covering glass.

[0164] After some minutes of boiling and stirring, the solution clarified and all the solid material dissolved. The boiling was maintained, and additional water was added when required, as to replace the water lost by boiling. After three hours of boiling, the solution was filtered while boiling on a Büchner funnel. Very small amounts of impurities were left in the filter. The filtrate was subsequently allowed to cool to room temperature, which resulted in growth of fine-powdered crystals of strontium aspartate trihydrate. Precipitation of the final product progressed in the filtrate within an hour. The product was filtered and dried at 110 °C in an oven for ½ hour followed by drying 12 hours in a dessicator over silica orange. Before analysis by x-ray crystallography and by FAAS, the salts were ground to fine powder by a mortar.

[0165] The total yield of strontium aspartate trihydrate was approximately 98% before recrystallisation, and the majority of impurities consisted of reminisces of the reagents and of strontium carbonate. This yield is significantly higher than the yield obtained by synthesis under conventional conditions where only 14 % was obtained (please see example 2). Thus the high temperature synthesis method as disclosed in this patent provides a significant gain in yield and a reduction in synthesis time, while resulting In a strontium aspartate salt of higher purity. The product was unambiguously identified as strontium aspartate trihydrate by x-ray crystallography and comparing the data to results of the Cambridge Crystallographic Database and information from H. Schmidbaur, P. Mikulcik & G. Müller (1990), Chem Ber. 123; 1599-1602 as depicted in figure 3 and table 4 and 5 herein.

[0166] Further improvements of the synthesis may include degassing by nitrogen or by argon of the water and of all aqueous solutions, which prevents contact to carbon dioxide that eventually may lead to formation of impurities of strontium carbonate. It follows that a person skilled in the art will easily be able to adapt the procedure to proceed under an inert gas atmosphere.

Example 6 (Reference)


Preparation of strontium malonate monohydrate by synthesis at 100°C



[0167] Initially, a suspension of malonic acid (white colored) is prepared by adding 100 mL of millipore water to 10.406 g (0.1 moles) of solid malonic acid (Fluka, , MW 104.06 g/mole, CAS no. 141-82-2, lot. no. 449503/1, filling code 44903076) in a 250 mL beaker. To this suspension was added 26.571 g (0.1 moles) of solid strontium hydroxide (Sigma Aldrich, Sr(OH)2*8H2O, MW 265.71, CAS no. 1311-10-0). Then, a magnetic stirring rod was added and the stirring and heating was started to the point of boiling of the suspension. The final suspension is also white colored and the stirring was sustained by maintaining a medium rotation rate of the stirring apparatus. In order to prevent carbon dioxide from entering the solution, the beaker was covered by a covering glass.

[0168] After some minutes of boiling and stirring, the solution clarified and all the solid material dissolved. The boiling was maintained, and additional water was added when required, as to replace the water lost by boiling. After three hours of boiling, the solution was filtered while boiling on a Büchner funnel. Very small amounts of impurities were left in the filter. The filtrate was subsequently allowed to cool to room temperature, which resulted in growth of fine-powdered crystals of strontium malonate. Precipitation of the final product progressed rapidly during filtration and the majority of the product was found in the filter (unheated). Only in rare instants, the precipitation progressed in the filtrate. The product was filtered and dried at 110°C in an oven for ½ hour followed by drying 12 hours in a dessicator over silica orange. Before analysis by x-ray crystallography and by FAAS, the salts were ground to fine powder by a mortar.

[0169] The total yield of strontium malonate was approximately 98% before recrystallisation, and the majority of impurities consisted of reminisces of the reagents and of strontium carbonate. The product was unambiguously identified as strontium malonate by x-ray crystallography and comparing the data to results of the Cambridge Crystallographic Database.

[0170] Further improvements of the synthesis may include degassing by nitrogen or by argon of the water and of all aqueous solutions, which prevents contact to carbon dioxide that eventually may lead to formation of impurities of strontium carbonate. It follows that a person skilled in the art will easily be able to adapt the procedure to proceed under an inert gas atmosphere.

Example 7


Methods of manufacture of water soluble strontium salts of dicarboxylic acids using temperatures above 100 °C



[0171] According to methods developed previously and described in examples 2 - 6, synthesis of strontium salts of dicarboxylic organic acids, and especially strontium salts of amino acids can be difficult to produce in larger scale (i.e. > 1 kg) due to low yields and difficulties in separating the desired reaction products from contaminants. Strontium salts of carbonate are of special concern as they will form as impurities when the reaction is occurring in atmospheric air containing normal levels of carbon dioxide. We have described in examples 4 - 6 that the total yield of the product when strontium salts of dicarboxylic acids are manufactured from the free acid form of the anion, and strontium hydroxide depends on temperature and on time of synthesis. In order for the reaction to reach completion, the mixture of the amino acid proper and strontium hydroxide needs boiling in water for three hours, allowing ample time for strontium in the reaction mixture to react with carbon dioxide in the air. In this example we disclose methods of improving the synthesis further by providing optimized reaction conditions, where temperature is increased above 100°C in a closed container, and where reaction times are significantly reduced.

[0172] The present example provides representative data from the optimization of conditions for synthesis of strontium glutamate in an autoclave system. Strontium glutamate is used as an example, but it the optimizations described in the example is also applicable for the synthesis of other strontium salts, where the exact reaction conditions can be optimized as disclosed in this example. The reaction temperatures must be maintained below the melting point or below the temperature of decomposition of the organic anion moiety of the desired strontium salt. As an example, malonic acid decomposes at 132-134 °C, and thus synthesis of strontium malonate must be performed at temperatures below 132°C.

[0173] Strontium L-glutamate was used as a model strontium compound In the optimisation experiments. The purity of the product was monitored by comparing to crystallographic data and by measuring the content of strontium. Ideally, the content of strontium is 25.7% in strontium L-glutamate hexahydrate, which is the product formed in these experiments. It follows that other soluble strontium salts may be prepared by similar methods with high yield and purity.

Experimental



[0174] Preparation of solutions: A suspension of glutamic acid (white coloured) is prepared by adding 100 mL of millipore water to 14.703 g (0.1 moles) of solid L-glutamic acid (Sigma Aldrich, C5H9NO4, MW 187.14 g/mole, CAS no. 142-47-2, lot. no. 426560/1, filling code 43003336) in a 250 mL beaker. To this suspension was added 22.257 g, 26.571 g or 31.885 (0.08 moles, 0.1 moles or 0.12 moles) of solid strontium hydroxide (Sigma Aldrich, Sr(OH)2*8H2O, MW 265.71, CAS no. 1311-10-0).

Optimisation experiments



[0175] After preparation of the salts, the nine optimisation experiments were performed according to the settings of table 9.
Table 9. Parameters and main results of the optimisation procedure for synthesis of strontium glutamate. The pressure was monitored but not used in the optimisation process. The strontium content (%Sr) was measured by FAAS but not used as quality parameter. The yield (%) was applied as the quality parameter.
Experiment no. Autoclave temperature (°C) Time of synthesis (min.) Base-acid ratio Total volume (ML) Autoclave pressure (bar) Yield% %SR (AAS)
1 125 15 0,8 50 1,55 94 25
2 124 30 1 75 1 112 22
3 124 60 1,2 100 1,6 121 21
4 127 15 0,8 100 1,2 118 22
5 132 30 1 50 1,55 120 25
6 132 60 1,2 75 1,6 50 22
7 134 15 0,8 75 1,65 108 24
8 134 30 1 100 1,65 76 14
9 132 60 1,2 50 1,65 82 24

Procedure



[0176] 

1. The calculated amount of acid was weighed and transferred to a bluecap autoclave bottle and the Millipore water was added. The bottle was closed and shaken, in order to obtain a finely grained suspension.

2 The calculated amount of strontium hydroxide octahydrate was weighed and added to the acid solution of (1) and the bottle was vigorously wortexed until all coarse lumps of material were transformed into fine-grained powder.

3 The bottle was placed in the autoclave and the temperature was set. While in the autoclave no additional stirring was carried out.

4 At t = 100° C the valve of the autoclave was closed and the timing was started.

5 During the autoclaving were monitored the actual temperature and the actual pressure.

6 After the time of autoclaving ended, the steam was let out, as soon as possible, with due respect to safety precautions.

7 At approx. 110° C the autoclave was opened and the solution was recovered. Again, the bottle was shook, as to obtain a high degree of mixing.

8 The solution was immediately filtered hot on a Büchner funnel after autoclaving, which left only traces of carbonate in the filter. The product precipitated from the solution during cooling to room temperature.

9 After precipitation, the product was filtered and dried in an oven for ½ an hour at 110° C.Then, it was dried in an dessicator over silica-gel orange. Finally, the product was ground to fine powder in a mortar.

10. The product was weighed after grinding and the total yield calculated.


Preparation of strontium malonate (Reference)



[0177] In order to confirm the applicability of the disclosed high temperature synthesis method for other strontium salts than strontium L-glutamate, strontium malonate was prepared. Basically the reaction conditions found for preparation of strontium L-glutamate was employed. A suspension of malonic acid (white coloured) is prepared by adding 100 mL of millipore water to 10.41 g (0.1 moles) of solid malonic acid (FLUKA 63290, MW 104.1) in a 250 mL beaker. To this suspension was added 22.257 g, 26.571 g or 31.885 (0.08 moles, 0.1 moles or 0.12 moles) of solid strontium hydroxide (Sigma Aldrich, Sr(OH)2*8H2O, MW 265.71, CAS no. 1311-10-0). The reaction procedure described above was follower, and the temperature was maintained below 130°C to avoid decomposition of malonic acid, while the reaction time was maintained at 15 min.

Content of strontium (% Sr):



[0178] A sample of 0.2 g was dissolved in 100 mL 0.1 M HNO3 prepared in Millipore water. This solution was further diluted by a factor of 500 by a solution of 1% KCI, and the content of strontium was determined by FAAS. The measurements were performed by using a Perkin-Elmer 2100 equipped with a hydrogen lamp for correction of the background signal. Strontium was measured at a slit with of 0.2 nm, the wavelength was 460.8 nm operated at an energy of 58 and a current of 8 mA.

X-ray crystallography



[0179] A second check of purity was performed by powder x-ray crystallography using a Huber G670 diffractometer. A characteristic diffractogram of the strontium glutamate is shown in fig. 5. An X-ray diffractogram of strontium malonate obtained by the high temperature synthesis method disclosed in the present example is shown in fig. 6. The double peak on the low angle side of the peak of maximum intensity observed in both figure 5 and 6 is an artefact of the instrument.

Results and discussion



[0180] In table 9, it is observed that some of the synthesis conditions resulted in relatively low yield and in strontium glutamate of low purity as apparent from the molar % of strontium in the reaction product. The product of experiment no. 8 was produced in relatively low yield, and it did not contain the expected 25.7% of strontium, which was also confirmed by the x-ray analysis. Despite this outlier, in general, the outcome of the optimisation experiments is close to the expected products. Incomplete reaction provides a product of too low content of strontium while formation of strontium carbonate during the synthesis gives a too high value of the strontium content. Conditions employed in experiments 1 and 5 gave the strontium content in best agreement with the expected value. Of notice, it is also apparent although the product of experiment no. 6 was produced in low yield; it contained an amount of strontium that corresponded to the expected value.

[0181] By studying the influence of the individual parameters on the total yield (table 9 and fig. 7), it becomes clear that temperature, time of autoclaving and base-acid ratio are important for the synthesis while total volume is less important. A yield higher than 100%, which is observed in experimental conditions 2, 3, 4, 5 and 7 originates from incomplete drying, but this effect is almost eliminated when the average values are considered, as in fig. 7. Thus, the maximum yield was obtained by using a high temperature (133 °C), a short time of autoclaving (15 min.) and a surplus of strontium hydroxide. Accordingly, temperature is more important than time but it compares in importance to the base-to-acid ratio. However, great care must exerted as to not exceed the temperature of decomposition in the synthesis of other strontium salts, which for, e.g., the malonate is 132-134 °C. A 10th experiment of control of optimisation was performed, as to confirm the maximum yield of the optimisation experiments.

[0182] Furthermore an additional experiment was performed to validate the applicability of the high temperature synthesis method for the preparation of other organic strontium salts than strontium L-glutamate. Strontium malonate was chosen, as this salt may be considered especially difficult to prepare under the high temperature conditions due to the low dissociation temperature of the malonic acid anion. However, as shown in figure 6, crystalline pure and well defined strontium malonate could easily be obtained. The crystal structure of the compound has not been completely resolved as it is a new structure not previously described, but the data shows that the high temperature method is likely to be applicable for many other organic strontium salts.

[0183] Further improvements of the synthesis include introduction of inert atmospheres to the synthesis environment, as well as degassing of all solutions by either nitrogen gas or by argon gas, as to reduce the formation of strontium carbonate.

Conclusion



[0184] The optimisation experiments show that it is possible to synthesize strontium glutamate in high yields by elevating the temperature to values above 100°C, and by using a short time (15 min.) in the autoclave. Also, a 20% surplus of strontium-hydroxide also improves the total yield without compromising the purity of the synthesized strontium salt. A slightly more vigorous drying than silica-gel orange should be applied to the drying procedure in order to obtain completely dried product. Examples of more potent drying agents are concentrated sulphuric acid or calcium oxide, but also conventional lyophilization or other mechanic treatments may be applicable for this procedure.

Example 8


Pharmacokinetic properties of dicarboxylic strontium salts



[0185] The aim of this experiment was to assess the bioavailability of dicarboxylic strontium salts compared with strontium chloride and strontium ranelate. The bioavailability was assessed by determination of serum strontium concentration at regular intervals over a 24 hour period and calculating AUC.

[0186] The experiment was performed with female SPF Wistar rats of the strain HanTac:WH (GALAS) from Taconic M&B A/S, Ejby, DK-4623 Lille Skensved, Denmark. At the start of the acclimatisation period, the rats were approximately 9 weeks old with a weight of approximately 200-250 g. The animals were housed in a room provided with filtered air at a temperature of 21°C ± 3°C and relative humidity of 55% ±15% and a ventilation system providing 10 air changes per hour. The room was illuminated to give a cycle of 12 hours light and 12 hours darkness. The rats were fed a complete pelleted rodent diet "Altromin 1314" (Chr. Petersen A/S, DK-4100 Ringsted, Denmark). The rats had free access to bottles with domestic quality drinking water acidified with hydrochloric acid to pH 2.5 in order to prevent microbial growth.

[0187] The rats were randomly allocated randomly in seven groups of 9 animals treated as indicated in the table below. The groups, dose levels, animal numbers were as listed in table 10:
Table 10: The 7 treatment groups of the pharmacokinetic experiment. The doses administered in the group are listed in the fist column, and salt, MW and Sr content in the middle columns.
Dose1 (mg /kg) Group Strontium salt MW % Sr Dose Equivalent1 (Amounts in mg) Animal No's
Vehicle Control Vehicle (0.5 % CMC) - - - 1-9
500 B Sr-ranelate (*7H2O) + 639.6 27.4 500 = 137 mg Sr++ 10-18
416 C SrCl2 (*6H2O) + 266.6 32.9 137 mg Sr++ = 416 . 19-27
533 D Sr-glutamate (*6H2O) 340.7 25.7 137 mg Sr++ = 533 28-36
427 E Sr-aspartate (*3H2O) 272.7 32.1 137 mg Sr++ = 427 37-45
484 F Sr-maleate (*6H2O) 309.7 28.3 137 mg Sr++ = 484 46-54
325 G Sr-malonate + (*1H2O) 207.7 42.2 137 mg Sr++ = 325 55-63
+ Reference compound
1Doses are adjusted to provide equimolar strontium dose as 500 mg/kg Strontium-ranelate (heptahydrate)(group B).


[0188] The test article (strontium salt) was given once by oral gavage according to the most recent body weight data. The control group was dosed with the vehicle alone (0.5% carboxy methyl cellulose, CMC). The vehicle was prepared with de-ionized water for all treatment groups including controls. The test substances (strontium salts) were solubilized/suspended in a volume corresponding to 5 ml/kg body weight. In order to keep the compounds in suspension, the formulations were kept on a magnetic stirrer before and during dosing.

Blood samples for toxicokinetics



[0189] On the day of treatment (Day 1), blood samples were taken from all animals. Blood samples were collected from 3 animals per group at the following time points: Pretreatment, and 30 min, 1, 1.5, 2, 4, 8 and 24 hours post-treatment, so that three animals from each group had samples taken at time 0, 1.5 and 6 hours, 3 other rats at time 0.5, 2, 8 hours and the remaining three animals in the group had samples taken at 1, 4 and 24 hours.

[0190] Approximately 0.5 - 0.6 ml blood was obtained at each time point from the orbital venous plexus into plain tubes for serum. The blood was kept at room temperature for 30 to 60 minutes and until centrifugation (10 min, 1270 G, +20 °C). The serum was transferred to Nunc cryotubes (Nunc, Denmark) and frozen at -18 °C for subsequent analysis of strontium content by graphite -furnace atomic-absorption spectrometry (GF-AAS).

Graphite-furnace atomic-absorption spectrometry (GF-AAS)



[0191] Concentrated HCl was added to the serum samples to a final concentration of 0.2% HCl and the samples were then subjected to analysis using a Perkin-Elmer 2100 equipped with a hydrogen lamp for correction of the background signal. Strontium was measured at a slit with of 0.2 nm, the wavelength was 460.8 nm operated at an energy of 58 and a current of 8 mA.

Results of the pharmacokinetic study of strontium salt absorption



[0192] In figure 8, the serum concentration measured in the six groups treated with strontium salts are plotted as a function of the time after administration of the compounds. It is apparent that administration of the strontium salts results in a rapid and highly significant increase in serum strontium concentrations. When comparing the pharmaco-kinetic properties of different salts, it is apparent that both the highly soluble strontium chloride as well as the relatively poorly soluble strontium ranelate (see example 3), is rapidly absorbed, reaching a maximum serum concentration after approximately 2 hours.

[0193] The di-carboxylic acids with higher solubility, and especially the strontium salts of the amino acids L-aspartate and L-glutamate reach the maximal serum concentration with a slower kinetic rate and, with maximal concentration reached after approximately 8 hours. Furthermore, the serum strontium concentration in the time interval from 0 - 8 hours after the administration of the test substance appears more stable, at least for some of the di-carboxylic acids such as the aspartate and malonate salts of strontium. This pattern of two distinct peaks of maximal serum concentration is also apparent in the group treated with strontium malonate. It is likely to indicate that the strontium ion is taken up by two distinct absorption mechanisms, and that the highly soluble strontium salts according to the present invention may have particular potential to exploit the biphasic nature of the strontium uptake mechanism, and thus proved an overall benefit apparent as higher bioavailability of the strontium.

[0194] When AUC calculations were performed the general course of the curves, as evidenced by average values in fig. 8, was best described by modelling the response/pharmacokinetic curves in a specially developed mathematical model. In the initial step, it assumes that the strontium is not metabolised but simply transferred from the stomach/upper digestive tract of the rat into epithelial cells by an active transport mechanism. Also without metabolism, the strontium ion is then transferred from the stomach/upper digestive tract where it is simultaneously released to the blood vessels. Only during the circulation of strontium through the veins, the strontium is dispersed and metabolised by the body tissue. This credible but simplified description, thus includes a two-step mechanism of absorption of ionic strontium after oral administrations of the strontium ions, identified by the two peaks of fig. 9 at t = 60 min and at t = 360 min. After the strontium dose was administered to the rats, a characteristic time of uptake was found as t = 12 min. The maximum content of strontium in the serum was observed after approx. 30 min. The characteristic time value of 12 min. is interpreted as the duration of strontium ions being taken up by the active transport mechanism from the intestinal lumen and secreted into circulation. The time of strontium transfer between the stomach and the blood vessels is initiated almost instantly, while the time of transfer between the guts and the blood vessels proceeds at a later stage that depends on the type of salt investigated. The malonate, in particular, exhibits a peak in the uptake-versus time from the guts to the blood vessels at t = 360 min., as seen in fig. 8. Thus, the time of body metabolism of the malonate is very long, as compared to that of the other salts. For all salts, however, the strontium content levels out after approx. 1750 min. (29 hours) and approaches the natural level corresponding to the pre-dose level.

[0195] The model calculations (not shown) were applied to the determination of the areas under the curve that are shown in table 11. The standard deviations of the AUC values correspond to the general uncertainty on the measurements of fig. 8, and their magnitude does not allow for a significant discrimination between the salts. The AUC values of the salts are much higher than the AUC value of the pre-dose samples.
Table 11. Determination of the area under the curve according (AUC) to the model calculations.
ANION OF Sr-SALT AUC mg / L·min STDDEV mg / L·min
α-ketoglutrate, 9000 1600
Aspartate 7000 1700
Chloride + 7300 2000
Glutamate 10100 3100
Malonate + 15000 8500
Pre-dose 168 67
Average 6800 5400
+ Reference compounds


[0196] These effects of delayed uptake of strontium and serum levels in a sustained level over longer time periods observed with strontium salts with di-carboxylic organic anions may enhance the pharmacological properties of the compounds. The delayed attainment of Cmax may be an advantage for the use of the strontium compound in the treatment of diseases and conditions affecting bone metabolism. In these cases it is often an advantage to administer the compound in the evening before bedtime, as this would allow the compound to act at night, when resorption of bone is occurring at the highest rate. Furthermore, the administration before bedtime minimizes the potential interference from calcium in the normal diet, as the pharmaceutical preparation of the strontium salt would be taken after the last meal. This is in contrast to administration during the day, where the calcium content of normal meals would have the potential to interfere and reduce the uptake of strontium. The gradual increase in serum strontium concentration over 4-8 hours after administration of the compound would comply well with evening administration of the compound and appears well suited to maximize the therapeutic effect of the strontium compound on bone metabolism.


Claims

1. A strontium salt having a water-solubility at room temperature in a range of from 1 g/l to 100 g/l for use in treatment of and/or prophylaxis of a cartilage and/or bone disease and/or conditions resulting in a dysregulation of cartilage and/or bone metabolism in a mammal, wherein the mammal is selected from a human female or male adult, adolescent or a child, and wherein the cartilage and/or bone disease and/or conditions resulting in a dysregulation of cartilage and/or bone metabolism is selected from osteoporosis, osteopenia, Paget's disease, osteolytic lesions produced by bone metastasis, bone loss due to sex steroid hormone deficiency, bone abnormalities caused by cancer therapeutics, immobilization-induced osteopenia or osteoporosis, glucocorticoid-induced osteopenia or osteoporosis, osteoporosis pseudoglioma syndrome, idiopathic juvenile osteoporosis or for the improvement of fracture healing after traumatic or atraumatic fracture,
and wherein the strontium salt is selected from the group consisting of strontium succinate, strontium glutamate, strontium aspartate, strontium maleate, strontium pyruvate and strontium alpha-ketoglutarate.
 
2. A strontium salt for use according to claim 1, wherein the strontium salt is strontium succinate.
 
3. A pharmaceutical composition comprising a strontium salt according to any of claims 1 to 2, together with one or more pharmaceutically acceptable excipients for use according to any of claims 1 to 2.
 
4. A pharmaceutical composition for use according to claim 3, wherein the pharmaceutical composition is in the form of tablets, capsules, sachets, powders, pellets, granules, granulates, mixtures, syrups, solutions, suspensions, or emulsions, for oral administration.
 


Ansprüche

1. Strontiumsalz mit einer Wasserlöslichkeit bei Raumtemperatur in einem Bereich von 1 g/l bis 100 g/l zur Verwendung in der Behandlung von und/oder der Prophylaxe einer Knorpel - und/oder Knochenerkrankung und/oder Zuständen, die zu einer Dysregulierung des Knorpel - und/oder Knochenstoffwechsels in einem Säugetier führen, wobei das Säugetier ausgewählt ist aus einem weiblichen Mensch oder männlichen Erwachsenen, Heranwachsenden oder einem Kind, und wobei die Knorpel- und/oder Knochenerkrankung und/oder die Zustände, die zu einer Dysregulierung des Knorpel- und/oder Knochenstoffwechsels führen ausgewählt ist aus Osteoporose, Osteopenie, Paget-Krankheit, durch Knochenmestastasen verursachte osteolytische Läsionen, Knochenverlust aufgrund von Steroid-Sexualhormon-Defizienz, durch Krebstherapeutika verursachte Knochenabnormalitäten, Immobilisierungs-induzierte Osteopenie oder Osteoporose, Glucocorticoid-induzierte Osteopenie oder Osteoporose, Osteoporose Pseudoglioma Syndrom, idiopathische juvenile Osteoporose oder zur Verbesserung der Frakturheilung nach traumatischer oder atraumatischer Fraktur, und wobei das Strontiumsalz ausgewählt ist aus der Gruppe bestehend aus Strontiumsuccinat, Strontiumglutamat, Strontiumaspartat, Strontiummaleat, Strontiumpyruvat und Strontiumalpha-Ketoglutarat,
 
2. Strontiumsalz zur Verwendung nach Anspruch 1, wobei das Strontiumsalz Strontiumsuccinat ist.
 
3. Pharmazeutische Zusammensetzung, umfassend ein Strontiumsalz nach einem der Ansprüche 1 oder 2, zusammen mit einem oder mehreren pharmazeutisch akzeptablen Hilfsstoffen zur Verwendung nach einem der Ansprüche 1 oder 2.
 
4. Pharmazeutische Zusammensetzung zur Verwendung nach Anspruch 3, wobei die pharmazeutische Zusammensetzung in der Form von Tabletten, Kapseln, Portionsbeuteln, Pulvern, Pellets, Granulae, Granulaten, Gemischen, Sirupen, Lösungen, Suspensionen oder Emulsionen für die orale Verabreichung vorliegt.
 


Revendications

1. Sel de strontium ayant une solubilité dans l'eau à température ambiante située dans une plage allant de 1 g/l à 100 g/l, pour une utilisation dans le traitement et/ou la prophylaxie d'une maladie du cartilage et/ou des os et/ou d'états ayant pour résultat une dérégulation du métabolisme du cartilage et/ou des os chez un mammifère, où le mammifère est choisi parmi des êtres humains de sexe féminin ou masculin adultes, adolescents ou enfants, et où la maladie du cartilage et/ou des os et/ou les états ayant pour résultat une dérégulation du métabolisme du cartilage et/ou des os sont choisis parmi l'ostéoporose, l'ostéopénie, la maladie de Paget, les lésions ostéolytiques produites par des métastases osseuses, une perte osseuse due à une insuffisance en hormone stéroïde sexuelle, les anomalies osseuses dues à un traitement anticancéreux, l'ostéopénie ou l'ostéoporose induite par une immobilisation, l'ostéopénie ou l'ostéoporose induite par un glucocorticoïde, le syndrome ostéoporose-pseudogliome, l'ostéoporose juvénile idiopathique, ou pour améliorer la consolidation d'une fracture après une fracture traumatique ou atraumatique,
lequel sel de strontium est choisi dans le groupe constitué par le succinate de strontium, le glutamate de strontium, l'aspartate de strontium, le maléate de strontium, le pyruvate de strontium et l'α-cétoglutarate de strontium.
 
2. Sel de strontium pour une utilisation selon la revendication 1, lequel sel de strontium est le succinate de strontium.
 
3. Composition pharmaceutique comprenant un sel de strontium selon l'une quelconque des revendications 1 à 2, conjointement avec un ou plusieurs excipients pharmaceutiquement acceptables, pour une utilisation selon l'une quelconque des revendications 1 à 2.
 
4. Composition pharmaceutique pour une utilisation selon la revendication 3, laquelle composition pharmaceutique est sous la forme de comprimés, capsules, sachets, poudres, pastilles, granules, granulés, mélanges, sirops, solutions, suspensions ou émulsions, pour une administration par voie orale.
 




Drawing
































Cited references

REFERENCES CITED IN THE DESCRIPTION



This list of references cited by the applicant is for the reader's convenience only. It does not form part of the European patent document. Even though great care has been taken in compiling the references, errors or omissions cannot be excluded and the EPO disclaims all liability in this regard.

Patent documents cited in the description




Non-patent literature cited in the description